201
|
Ma L, Jongbloets BC, Xiong WH, Melander JB, Qin M, Lameyer TJ, Harrison MF, Zemelman BV, Mao T, Zhong H. A Highly Sensitive A-Kinase Activity Reporter for Imaging Neuromodulatory Events in Awake Mice. Neuron 2018; 99:665-679.e5. [PMID: 30100256 PMCID: PMC6152931 DOI: 10.1016/j.neuron.2018.07.020] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 06/01/2018] [Accepted: 07/11/2018] [Indexed: 01/01/2023]
Abstract
Neuromodulation imposes powerful control over brain function, and cAMP-dependent protein kinase (PKA) is a central downstream mediator of multiple neuromodulators. Although genetically encoded PKA sensors have been developed, single-cell imaging of PKA activity in living mice has not been established. Here, we used two-photon fluorescence lifetime imaging microscopy (2pFLIM) to visualize genetically encoded PKA sensors in response to the neuromodulators norepinephrine and dopamine. We screened available PKA sensors for 2pFLIM and further developed a variant (named tAKARα) with increased sensitivity and a broadened dynamic range. This sensor allowed detection of PKA activation by norepinephrine at physiologically relevant concentrations and kinetics, and by optogenetically released dopamine. In vivo longitudinal 2pFLIM imaging of tAKARα tracked bidirectional PKA activities in individual neurons in awake mice and revealed neuromodulatory PKA events that were associated with wakefulness, pharmacological manipulation, and locomotion. This new sensor combined with 2pFLIM will enable interrogation of neuromodulation-induced PKA signaling in awake animals. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Lei Ma
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Bart C Jongbloets
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Wei-Hong Xiong
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Joshua B Melander
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Maozhen Qin
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Tess J Lameyer
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Madeleine F Harrison
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX 78712, USA
| | - Boris V Zemelman
- Center for Learning and Memory, The University of Texas at Austin, Austin, TX 78712, USA
| | - Tianyi Mao
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Haining Zhong
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
202
|
Shao D, Villet O, Zhang Z, Choi SW, Yan J, Ritterhoff J, Gu H, Djukovic D, Christodoulou D, Kolwicz SC, Raftery D, Tian R. Glucose promotes cell growth by suppressing branched-chain amino acid degradation. Nat Commun 2018; 9:2935. [PMID: 30050148 PMCID: PMC6062555 DOI: 10.1038/s41467-018-05362-7] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 06/13/2018] [Indexed: 01/13/2023] Open
Abstract
Glucose and branched-chain amino acids (BCAAs) are essential nutrients and key determinants of cell growth and stress responses. High BCAA level inhibits glucose metabolism but reciprocal regulation of BCAA metabolism by glucose has not been demonstrated. Here we show that glucose suppresses BCAA catabolism in cardiomyocytes to promote hypertrophic response. High glucose inhibits CREB stimulated KLF15 transcription resulting in downregulation of enzymes in the BCAA catabolism pathway. Accumulation of BCAA through the glucose-KLF15-BCAA degradation axis is required for the activation of mTOR signaling during the hypertrophic growth of cardiomyocytes. Restoration of KLF15 prevents cardiac hypertrophy in response to pressure overload in wildtype mice but not in mutant mice deficient of BCAA degradation gene. Thus, regulation of KLF15 transcription by glucose is critical for the glucose-BCAA circuit which controls a cascade of obligatory metabolic responses previously unrecognized for cell growth.
Collapse
Affiliation(s)
- Dan Shao
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, 98109, USA
| | - Outi Villet
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, 98109, USA
| | - Zhen Zhang
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, 98109, USA
| | - Sung Won Choi
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, 98109, USA
| | - Jie Yan
- Department of Medicine, NMR Laboratory of Physiological Chemistry, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Julia Ritterhoff
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, 98109, USA
| | - Haiwei Gu
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, 98109, USA
| | - Danijel Djukovic
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, 98109, USA
| | - Danos Christodoulou
- Department of Medicine, NMR Laboratory of Physiological Chemistry, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Stephen C Kolwicz
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, 98109, USA
| | - Daniel Raftery
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, 98109, USA
- Fred Hutchinson Cancer Research Center, 1100 Fairview Ave, Seattle, WA, 98109, USA
| | - Rong Tian
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, WA, 98109, USA.
| |
Collapse
|
203
|
Considerations for the use of virally delivered genetic tools for in-vivo circuit analysis and behavior in mutant mice: a practical guide to optogenetics. Behav Pharmacol 2018; 28:598-609. [PMID: 29099403 DOI: 10.1097/fbp.0000000000000361] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Optogenetics was the method of the year in 2010 according to Nature Neuroscience. Since then, this method has become widespread, the use of virally delivered genetic tools has extended to other fields such as pharmacogenetics, and optogenetic techniques have become frequently applied in genetically manipulated animals for in-vivo circuit analysis and behavioral studies. However, several issues should be taken into consideration when planning such experiments. We aimed to summarize the critical points concerning optogenetic manipulation of a specific brain area in mutant mice. First, the appropriate vector should be chosen to allow optimal optogenetic manipulation. Adeno-associated viral vectors are the most common carriers with different available serotypes. Light-sensitive channels are available in many forms, and the expression of the delivered genetic material can be influenced in many ways. Second, selecting the adequate stimulation protocol is also essential. The pattern, intensity, and timing could be determinative parameters. Third, the mutant strain might have a phenotype that influences the observed behavior. In conclusion, detailed preliminary experiments and numerous control groups are required to choose the best vector and stimulation protocol and to ensure that the mutant animals do not have a specific phenotype that can influence the examined behavior.
Collapse
|
204
|
Bedbrook CN, Deverman BE, Gradinaru V. Viral Strategies for Targeting the Central and Peripheral Nervous Systems. Annu Rev Neurosci 2018; 41:323-348. [DOI: 10.1146/annurev-neuro-080317-062048] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recombinant viruses allow for targeted transgene expression in specific cell populations throughout the nervous system. The adeno-associated virus (AAV) is among the most commonly used viruses for neuroscience research. Recombinant AAVs (rAAVs) are highly versatile and can package most cargo composed of desired genes within the capsid's ∼5-kb carrying capacity. Numerous regulatory elements and intersectional strategies have been validated in rAAVs to enable cell type–specific expression. rAAVs can be delivered to specific neuronal populations or globally throughout the animal. The AAV capsids have natural cell type or tissue tropism and trafficking that can be modified for increased specificity. Here, we describe recently engineered AAV capsids and associated cargo that have extended the utility of AAVs in targeting molecularly defined neurons throughout the nervous system, which will further facilitate neuronal circuit interrogation and discovery.
Collapse
Affiliation(s)
- Claire N. Bedbrook
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Benjamin E. Deverman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| |
Collapse
|
205
|
Wu Y, Jiang L, Geng H, Yang T, Han Z, He X, Lin K, Xu F. A Recombinant Baculovirus Efficiently Generates Recombinant Adeno-Associated Virus Vectors in Cultured Insect Cells and Larvae. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 10:38-47. [PMID: 29988889 PMCID: PMC6034586 DOI: 10.1016/j.omtm.2018.05.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 05/21/2018] [Indexed: 12/12/2022]
Abstract
Current large-scale recombinant adeno-associated virus (rAAV) production systems based on the baculovirus expression vector (BEV) remain complicated and cost-intensive, and they lack versatility and flexibility. Here we present a novel recombinant baculovirus integrated with all packaging elements for the production of rAAV. To optimize BEV construction, ribosome leaky-scanning mechanism was used to express AAV Rep and Cap proteins downstream of the PH and P10 promoters in the pFast.Bac.Dual vector, respectively, and the rAAV genome was inserted between the two promoters. The yields of rAAV2, rAAV8, and rAAV9 derived from the BEV-infected Sf9 cells exceeded 105 vector genomes (VG) per cell. The BEV was shown to be stable and showed no apparent decrease of rAAV yield after at least four serial passages. The rAAVs derived from the new Bac system displayed high-quality and high-transduction activity. Additionally, rAAV2 could be efficiently generated from BEV-infected beet armyworm larvae at a per-larvae yield of 2.75 ± 1.66 × 1010 VG. The rAAV2 derived from larvae showed a structure similar to the rAAV2 derived from HEK293 cells, and it also displayed high-transduction activity. In summary, the novel BEV is ideally suitable for large-scale rAAV production. Further, this study exploits a potential cost-efficient platform for rAAV production in insect larvae.
Collapse
Affiliation(s)
- Yang Wu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.,Brain Research Center, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China
| | - Liangyu Jiang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.,Brain Research Center, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China
| | - Hao Geng
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.,Brain Research Center, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China
| | - Tian Yang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.,Brain Research Center, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China
| | - Zengpeng Han
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.,Brain Research Center, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China
| | - Xiaobing He
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.,Brain Research Center, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China
| | - Kunzhang Lin
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.,Brain Research Center, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China
| | - Fuqiang Xu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.,Brain Research Center, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
206
|
Serum exosomes mediate delivery of arginase 1 as a novel mechanism for endothelial dysfunction in diabetes. Proc Natl Acad Sci U S A 2018; 115:E6927-E6936. [PMID: 29967177 DOI: 10.1073/pnas.1721521115] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Exosomes, abundant in blood, deliver various molecules to recipient cells. Endothelial cells are directly exposed to circulating substances. However, how endothelial cells respond to serum exosomes (SExos) and the implications in diabetes-associated vasculopathy have never been explored. In the present study, we showed that SExos from diabetic db/db mice (db/db SExos) were taken up by aortic endothelial cells, which severely impaired endothelial function in nondiabetic db/m+ mice. The exosomal proteins, rather than RNAs, mostly account for db/db SExos-induced endothelial dysfunction. Comparative proteomics analysis showed significant increase of arginase 1 in db/db SExos. Silence or overexpression of arginase 1 confirmed its essential role in db/db SExos-induced endothelial dysfunction. This study is a demonstration that SExos deliver arginase 1 protein to endothelial cells, representing a cellular mechanism during development of diabetic endothelial dysfunction. The results expand the scope of blood-borne substances that monitor vascular homeostasis.
Collapse
|
207
|
Abstract
De novo protein synthesis is critical for memory formation. We found that protein synthesis during acquisition is transiently required for contextual memory formation. We identified one candidate gene, Nrgn (encoding protein neurogranin, Ng) with enhanced translation upon novel-context exposure, and found that experience-dependent translation of Ng in the hippocampus is required for contextual memory formation. Furthermore, fragile-X mental retardation protein interacts with the 3′UTR of the Nrgn mRNA, which is required for activity-dependent translation of Ng in the synaptic compartment and contextual memory formation. Together, these results indicate that experience-dependent and acute translation of Ng in the hippocampus during memory acquisition enables durable context memory encoding. Experience induces de novo protein synthesis in the brain and protein synthesis is required for long-term memory. It is important to define the critical temporal window of protein synthesis and identify newly synthesized proteins required for memory formation. Using a behavioral paradigm that temporally separates the contextual exposure from the association with fear, we found that protein synthesis during the transient window of context exposure is required for contextual memory formation. Among an array of putative activity-dependent translational neuronal targets tested, we identified one candidate, a schizophrenia-associated candidate mRNA, neurogranin (Ng, encoded by the Nrgn gene) responding to novel-context exposure. The Ng mRNA was recruited to the actively translating mRNA pool upon novel-context exposure, and its protein levels were rapidly increased in the hippocampus. By specifically blocking activity-dependent translation of Ng using virus-mediated molecular perturbation, we show that experience-dependent translation of Ng in the hippocampus is required for contextual memory formation. We further interrogated the molecular mechanism underlying the experience-dependent translation of Ng, and found that fragile-X mental retardation protein (FMRP) interacts with the 3′UTR of the Nrgn mRNA and is required for activity-dependent translation of Ng in the synaptic compartment and contextual memory formation. Our results reveal that FMRP-mediated, experience-dependent, rapid enhancement of Ng translation in the hippocampus during the memory acquisition enables durable context memory encoding.
Collapse
|
208
|
Chamberlain K, Riyad JM, Garnett T, Kohlbrenner E, Mookerjee A, Elmastour F, Benard L, Chen J, VandenDriessche T, Chuah MK, Marian AJ, Hajjar RJ, Gurha P, Weber T. A Calsequestrin Cis-Regulatory Motif Coupled to a Cardiac Troponin T Promoter Improves Cardiac Adeno-Associated Virus Serotype 9 Transduction Specificity. Hum Gene Ther 2018; 29:927-937. [PMID: 29641321 DOI: 10.1089/hum.2017.188] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Adeno-associated virus serotype 9 (AAV9) is an efficient vector for gene transfer to the myocardium. However, the use of ubiquitous promoters, such as the cytomegalovirus (CMV) promoter, can result in expression of the transgene in organs other than the heart. This study tested if the efficiency and specificity of cardiac transcription from a chicken cardiac troponin T (TnT) promoter could be further increased by incorporating a cardiomyocyte-specific transcriptional cis-regulatory motif from human calsequestrin 2 (CS-CRM4) into the expression cassette (Enh.TnT). The efficiency of luciferase expression from the TnT and Enh.TnT constructs was compared to expression of luciferase under the control of the CMV promoter in both adult and neonatal mice. Overall, expression levels of luciferase in the heart were similar in mice injected with AAV9.TnT.Luc, AAV9.Enh.TnT.Luc and AAV9.CMV.Luc. In contrast, expression levels of luciferase activity in nontarget organs, including the liver and muscle, was lower in mice injected with the AAV9.TnT.Luc compared to AAV9.CMV.Luc and was negligible with AAV9.Enh.TnT. In neonates, in organs other than the heart, luciferase expression levels were too low to be quantified for all constructs. Taken together, the data show that the AAV9 Enh.TnT constructs drives high levels of expression of the transgene in the myocardium, with insignificant expression in other organs. These properties reduce the risks associated with the AAV9-mediated expression of the therapeutic protein of interest in nontarget organs. The excellent cardiac specificity should allow for the use of higher vector doses than are currently used, which might be essential to achieve the levels of transgene expression necessary for therapeutic benefits. Taken together, the findings suggest that the Enh.TnT transcription unit is a potentially attractive tool for clinical cardiac gene therapy in adults.
Collapse
Affiliation(s)
- Kyle Chamberlain
- 1 Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Jalish M Riyad
- 1 Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Tyrone Garnett
- 3 Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, and Texas Heart Institute, Houston, Texas
| | - Erik Kohlbrenner
- 1 Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Ananda Mookerjee
- 1 Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Firas Elmastour
- 1 Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Ludovic Benard
- 1 Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Jiqiu Chen
- 1 Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Thierry VandenDriessche
- 4 Department of Gene Therapy and Regenerative Medicine, Free University of Brussels (VUB), Brussels, Belgium .,5 Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven , Leuven, Belgium
| | - Marinee K Chuah
- 4 Department of Gene Therapy and Regenerative Medicine, Free University of Brussels (VUB), Brussels, Belgium .,5 Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven , Leuven, Belgium
| | - Ali J Marian
- 3 Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, and Texas Heart Institute, Houston, Texas
| | - Roger J Hajjar
- 1 Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Priyatansh Gurha
- 3 Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, and Texas Heart Institute, Houston, Texas
| | - Thomas Weber
- 1 Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York City, New York.,2 Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York City, New York
| |
Collapse
|
209
|
Moreno AM, Fu X, Zhu J, Katrekar D, Shih YRV, Marlett J, Cabotaje J, Tat J, Naughton J, Lisowski L, Varghese S, Zhang K, Mali P. In Situ Gene Therapy via AAV-CRISPR-Cas9-Mediated Targeted Gene Regulation. Mol Ther 2018; 26:1818-1827. [PMID: 29754775 DOI: 10.1016/j.ymthe.2018.04.017] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 04/12/2018] [Accepted: 04/18/2018] [Indexed: 12/18/2022] Open
Abstract
Development of efficacious in vivo delivery platforms for CRISPR-Cas9-based epigenome engineering will be critical to enable the ability to target human diseases without permanent modification of the genome. Toward this, we utilized split-Cas9 systems to develop a modular adeno-associated viral (AAV) vector platform for CRISPR-Cas9 delivery to enable the full spectrum of targeted in situ gene regulation functionalities, demonstrating robust transcriptional repression (up to 80%) and activation (up to 6-fold) of target genes in cell culture and mice. We also applied our platform for targeted in vivo gene-repression-mediated gene therapy for retinitis pigmentosa. Specifically, we engineered targeted repression of Nrl, a master regulator of rod photoreceptor determination, and demonstrated Nrl knockdown mediates in situ reprogramming of rod cells into cone-like cells that are resistant to retinitis pigmentosa-specific mutations, with concomitant prevention of secondary cone loss. Furthermore, we benchmarked our results from Nrl knockdown with those from in vivo Nrl knockout via gene editing. Taken together, our AAV-CRISPR-Cas9 platform for in vivo epigenome engineering enables a robust approach to target disease in a genomically scarless and potentially reversible manner.
Collapse
Affiliation(s)
- Ana M Moreno
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Xin Fu
- Shiley Eye Institute, Institute for Engineering in Medicine, Institute for Genomics Medicine, University of California, San Diego, San Diego, CA, USA; Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jie Zhu
- Shiley Eye Institute, Institute for Engineering in Medicine, Institute for Genomics Medicine, University of California, San Diego, San Diego, CA, USA; Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Dhruva Katrekar
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Yu-Ru V Shih
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - John Marlett
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jessica Cabotaje
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Jasmine Tat
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - John Naughton
- Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Leszek Lisowski
- Translational Vectorology Group, Children's Medical Research Institute, University of Sydney, Sydney, NSW 2006, Australia; Military Institute of Hygiene and Epidemiology, The Biological Threats Identification and Countermeasure Centre, 24-100 Puławy, Poland
| | - Shyni Varghese
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA; Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Kang Zhang
- Shiley Eye Institute, Institute for Engineering in Medicine, Institute for Genomics Medicine, University of California, San Diego, San Diego, CA, USA; Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China; Veterans Administration Healthcare System, San Diego, CA, USA.
| | - Prashant Mali
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
210
|
Santacruz L, Arciniegas AJL, Darrabie M, Mantilla JG, Baron RM, Bowles DE, Mishra R, Jacobs DO. Hypoxia decreases creatine uptake in cardiomyocytes, while creatine supplementation enhances HIF activation. Physiol Rep 2018; 5:5/16/e13382. [PMID: 28821596 PMCID: PMC5582266 DOI: 10.14814/phy2.13382] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 07/18/2017] [Indexed: 12/23/2022] Open
Abstract
Creatine (Cr), phosphocreatine (PCr), and creatine kinases (CK) comprise an energy shuttle linking ATP production in mitochondria with cellular consumption sites. Myocytes cannot synthesize Cr: these cells depend on uptake across the cell membrane by a specialized creatine transporter (CrT) to maintain intracellular Cr levels. Hypoxia interferes with energy metabolism, including the activity of the creatine energy shuttle, and therefore affects intracellular ATP and PCr levels. Here, we report that exposing cultured cardiomyocytes to low oxygen levels rapidly diminishes Cr transport by decreasing Vmax and Km. Pharmacological activation of AMP‐activated kinase (AMPK) abrogated the reduction in Cr transport caused by hypoxia. Cr supplementation increases ATP and PCr content in cardiomyocytes subjected to hypoxia, while also significantly augmenting the cellular adaptive response to hypoxia mediated by HIF‐1 activation. Our results indicate that: (1) hypoxia reduces Cr transport in cardiomyocytes in culture, (2) the cytoprotective effects of Cr supplementation are related to enhanced adaptive physiological responses to hypoxia mediated by HIF‐1, and (3) Cr supplementation increases the cellular ATP and PCr content in RNCMs exposed to hypoxia.
Collapse
Affiliation(s)
- Lucia Santacruz
- Department of Molecular Biology and Biochemistry, The University of Texas Medical Branch, Galveston, Texas .,Department of Natural Sciences, Bowie State University, Bowie, Maryland
| | - Antonio Jose Luis Arciniegas
- Department of Medicine, Pulmonary and Critical Care Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | - Rebecca M Baron
- Department of Medicine, Pulmonary and Critical Care Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Dawn E Bowles
- Duke University Medical Center, Durham, North Carolina
| | | | - Danny O Jacobs
- Department of Surgery, The University of Texas Medical Branch, Galveston, Texas.,Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
211
|
Selective elimination of long INterspersed element-1 expressing tumour cells by targeted expression of the HSV-TK suicide gene. Oncotarget 2018; 8:38239-38250. [PMID: 28415677 PMCID: PMC5503529 DOI: 10.18632/oncotarget.16013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 03/02/2017] [Indexed: 12/31/2022] Open
Abstract
In gene therapy, effective and selective suicide gene expression is crucial. We exploited the endogenous Long INterspersed Element-1 (L1) machinery often reactivated in human cancers to integrate the Herpes Simplex Virus Thymidine Kinase (HSV-TK) suicide gene selectively into the genome of cancer cells. We developed a plasmid-based system directing HSV-TK expression only when reverse transcribed and integrated in the host genome via the endogenous L1 ORF1/2 proteins and an Alu element. Delivery of these new constructs into cells followed by Ganciclovir (GCV) treatment selectively induced mortality of L1 ORF1/2 protein expressing cancer cells, but had no effect on primary cells that do not express L1 ORF1/2. This novel strategy for selective targeting of tumour cells provides high tolerability as the HSV-TK gene cannot be expressed without reverse transcription and integration, and high selectivity as these processes take place only in cancer cells expressing high levels of functional L1 ORF1/2.
Collapse
|
212
|
Petit L, Ma S, Cheng SY, Gao G, Punzo C. Rod Outer Segment Development Influences AAV-Mediated Photoreceptor Transduction After Subretinal Injection. Hum Gene Ther 2018; 28:464-481. [PMID: 28510482 PMCID: PMC5488363 DOI: 10.1089/hum.2017.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Vectors based on the adeno-associated virus (AAV) are currently the preferred tools for delivering genes to photoreceptors (PR) in small and large animals. AAVs have been applied successfully in various models of PR dystrophies. However, unknown barriers still limit AAV's efficient application in several forms of severe PR degenerations due to insufficient transgene expression and/or treated cells at the time of injection. Optimizations of PR gene therapy strategies will likely benefit from the identification of the cellular factors that influence PR transduction. Interestingly, recent studies have shown that the AAV transduction profile of PRs differs significantly between neonatal and adult mouse retinas after subretinal injection. This phenomenon may provide clues to identify host factors that influence the efficiency of AAV-mediated PR transduction. This study demonstrates that rod outer segments are critical modulators of efficient AAV-mediated rod transduction. During retinal development, rod transduction correlated temporally and spatially with the differentiation order of PRs when vectors were introduced subretinally but not when introduced intravitreally. All subretinally injected vectors had an initial preference to transduce cones in the absence of formed rod outer segments and then displayed a preference for rods as the cells matured, independently of the expression cassette or AAV serotype. Consistent with this observation, altered development of rod outer segments was associated with a strong reduction of rod transduction and an increase in the percentage of transduced cones by 2- to 2.8-fold. A similar increase of cone transduction was observed in the adult retinal degeneration 1 (rd1) retina compared to wild-type mice. These results suggest that the loss of rod outer segments in diseased retinas could markedly affect gene transfer efficiency of AAV vectors by limiting the ability of AAVs to infect dying rods efficiently. This information could be exploited for the development of more efficient AAV-based PR gene delivery procedures.
Collapse
Affiliation(s)
- Lolita Petit
- 1 Department of Ophthalmology and Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Shan Ma
- 1 Department of Ophthalmology and Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Shun-Yun Cheng
- 1 Department of Ophthalmology and Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Guangping Gao
- 3 Department of Microbiology and Physiological Systems and Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Claudio Punzo
- 1 Department of Ophthalmology and Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts.,2 Department of Neurobiology, University of Massachusetts Medical School , Worcester, Massachusetts
| |
Collapse
|
213
|
Durost PA, Aryee KE, Manzoor F, Tisch RM, Mueller C, Jurczyk A, Shultz LD, Brehm MA. Gene Therapy with an Adeno-Associated Viral Vector Expressing Human Interleukin-2 Alters Immune System Homeostasis in Humanized Mice. Hum Gene Ther 2018; 29:352-365. [DOI: 10.1089/hum.2017.072] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Philip A. Durost
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Ken-Edwin Aryee
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Fatima Manzoor
- Department of Immunology and Microbiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Roland M. Tisch
- Department of Immunology and Microbiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Christian Mueller
- Department of Pediatrics and Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Agata Jurczyk
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | | | - Michael A. Brehm
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| |
Collapse
|
214
|
Robert MA, Nassoury N, Chahal PS, Venne MH, Racine T, Qiu X, Kobinger G, Kamen A, Gilbert R, Gaillet B. Gene Transfer of ZMapp Antibodies Mediated by Recombinant Adeno-Associated Virus Protects Against Ebola Infections. Hum Gene Ther 2018; 29:452-466. [PMID: 29179602 DOI: 10.1089/hum.2017.101] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Vectored delivery of the ZMapp antibody cocktail (c2G4, c4G7, and c13C6) by using recombinant adeno-associated viruses (rAAVs) could be useful for preventive immunization against Ebola virus infections because rAAVs can generate long-term antibody expression. Three rAAVs (serotype 9) encoding chimeric ZMapp antibodies were produced by triple-plasmid transfection up to 10 L-scale in WAVE bioreactors using HEK293 cells grown in suspension/serum-free conditions. Efficacy of AAV-c2G4 via intravenous (i.v.), intramuscular (i.m.), and intranasal (i.n.) routes of administration was evaluated in mice with two different doses of 2.7 × 1010 and 13.0 × 1010 vector genomes (vg). The best protective efficacies after Ebola challenge were obtained with the i.v. and i.m. routes. Serum concentrations of ZMapp antibodies positively correlated with survivability. Efficacy of the rAAV-ZMapp cocktail was then evaluated at a higher dose of 30.0 × 1010 vg. It conferred a more robust protection (90% i.v. and 60% i.m.) than rAAV-c4G7 (30%) and rAAV-c13C6 (70%), both administered separately at the same dose. Delivery of rAAV-c2G4 alone achieved up to 100% protection (100% i.v. and 90% i.m.) at the same dose. In conclusion, the preventive treatment was effective in mice. However, no advantage was observed for using the rAAV-ZMapp cocktail in comparison to the utilization of the single rAAV-c2G4.
Collapse
Affiliation(s)
- Marc-André Robert
- 1 Department of Chemical Engineering, Université Laval , Quebec, Canada .,2 National Research Council Canada , Montreal, Canada
| | | | | | | | - Trina Racine
- 3 Infectious Disease Research Center, Université Laval , Quebec, Canada .,4 Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Xiangguo Qiu
- 4 Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada .,5 Department of Medical Microbiology, McGill University , Montreal, Canada
| | - Gary Kobinger
- 3 Infectious Disease Research Center, Université Laval , Quebec, Canada .,6 Department of Laboratory Medicine, University of Pennsylvania , Philadelphia, Pennsylvania.,7 Department of Microbiology, University of Manitoba , Winnipeg, Canada
| | - Amine Kamen
- 8 Department of Bioengineering, McGill University , Montreal, Canada
| | | | - Bruno Gaillet
- 1 Department of Chemical Engineering, Université Laval , Quebec, Canada
| |
Collapse
|
215
|
Katrekar D, Moreno AM, Chen G, Worlikar A, Mali P. Oligonucleotide conjugated multi-functional adeno-associated viruses. Sci Rep 2018; 8:3589. [PMID: 29483550 PMCID: PMC5827683 DOI: 10.1038/s41598-018-21742-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 02/07/2018] [Indexed: 02/06/2023] Open
Abstract
Recombinant adeno-associated viruses (AAVs) are among the most commonly used vehicles for in vivo gene delivery. However, their tropism is limited, and additionally their efficacy can be negatively affected by prevalence of neutralizing antibodies in sera. Methodologies to systematically engineer AAV capsid properties would thus be of great relevance. In this regard, we develop here multi-functional AAVs by engineering precision tethering of oligonucleotides onto the AAV surface, and thereby enabling a spectrum of nucleic-acid programmable functionalities. Towards this, we engineered genetically encoded incorporation of unnatural amino acids (UAA) bearing bio-orthogonal chemical handles onto capsid proteins. Via these we enabled site-specific coupling of oligonucleotides onto the AAV capsid surface using facile click chemistry. The resulting oligo-AAVs could be sequence specifically labeled, and also patterned in 2D using DNA array substrates. Additionally, we utilized these oligo conjugations to engineer viral shielding by lipid-based cloaks that efficaciously protected the AAV particles from neutralizing serum. We confirmed these 'cloaked AAVs' retained full functionality via their ability to transduce a range of cell types, and also enable robust delivery of CRISPR-Cas9 effectors. Taken together, we anticipate this programmable oligo-AAV system will have broad utility in synthetic biology and AAV engineering applications.
Collapse
Affiliation(s)
- Dhruva Katrekar
- Department of Bioengineering, University of California, San Diego, CA, USA
| | - Ana M Moreno
- Department of Bioengineering, University of California, San Diego, CA, USA
| | - Genghao Chen
- Department of Bioengineering, University of California, San Diego, CA, USA
| | - Atharv Worlikar
- Department of Bioengineering, University of California, San Diego, CA, USA
| | - Prashant Mali
- Department of Bioengineering, University of California, San Diego, CA, USA.
| |
Collapse
|
216
|
Michalakis S, Schön C, Becirovic E, Biel M. Gene therapy for achromatopsia. J Gene Med 2018; 19. [PMID: 28095637 DOI: 10.1002/jgm.2944] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 01/13/2017] [Accepted: 01/13/2017] [Indexed: 02/02/2023] Open
Abstract
The present review summarizes the current status of achromatopsia (ACHM) gene therapy-related research activities and provides an outlook for their clinical application. ACHM is an inherited eye disease characterized by a congenital absence of cone photoreceptor function. As a consequence, ACHM is associated with strongly impaired daylight vision, photophobia, nystagmus and a lack of color discrimination. Currently, six genes have been linked to ACHM. Up to 80% of the patients carry mutations in the genes CNGA3 and CNGB3 encoding the two subunits of the cone cyclic nucleotide-gated channel. Various animal models of the disease have been established and their characterization has helped to increase our understanding of the pathophysiology associated with ACHM. With the advent of adeno-associated virus vectors as valuable gene delivery tools for retinal photoreceptors, a number of promising gene supplementation therapy programs have been initiated. In recent years, huge progress has been made towards bringing a curative treatment for ACHM into clinics. The first clinical trials are ongoing or will be launched soon and are expected to contribute important data on the safety and efficacy of ACHM gene supplementation therapy.
Collapse
Affiliation(s)
- Stylianos Michalakis
- Center for Integrated Protein Science Munich CiPSM and Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christian Schön
- Center for Integrated Protein Science Munich CiPSM and Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Elvir Becirovic
- Center for Integrated Protein Science Munich CiPSM and Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Martin Biel
- Center for Integrated Protein Science Munich CiPSM and Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
217
|
Abstract
Genome editing via homologous recombination (HR) (gene targeting) in human hematopoietic stem cells (HSCs) has the power to reveal gene-function relationships and potentially transform curative hematological gene and cell therapies. However, there are no comprehensive and reproducible protocols for targeting HSCs for HR. Herein, we provide a detailed protocol for the production, enrichment, and in vitro and in vivo analyses of HR-targeted HSCs by combining CRISPR/Cas9 technology with the use of rAAV6 and flow cytometry. Using this protocol, researchers can introduce single-nucleotide changes into the genome or longer gene cassettes with the precision of genome editing. Along with our troubleshooting and optimization guidelines, researchers can use this protocol to streamline HSC genome editing at any locus of interest. The in vitro HSC-targeting protocol and analyses can be completed in 3 weeks, and the long-term in vivo HSC engraftment analyses in immunodeficient mice can be achieved in 16 weeks. This protocol enables manipulation of genes for investigation of gene functions during hematopoiesis, as well as for the correction of genetic mutations in HSC transplantation-based therapies for diseases such as sickle cell disease, β-thalassemia, and primary immunodeficiencies.
Collapse
|
218
|
Julien L, Chassagne J, Peccate C, Lorain S, Piétri-Rouxel F, Danos O, Benkhelifa-Ziyyat S. RFX1 and RFX3 Transcription Factors Interact with the D Sequence of Adeno-Associated Virus Inverted Terminal Repeat and Regulate AAV Transduction. Sci Rep 2018; 8:210. [PMID: 29317724 PMCID: PMC5760533 DOI: 10.1038/s41598-017-18604-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/13/2017] [Indexed: 12/31/2022] Open
Abstract
Adeno-associated virus (AAV) transduction efficiency depends on the way in which cellular proteins process viral genomes in the nucleus. In this study, we have investigated the binding of nuclear proteins to the double stranded D (dsD) sequence of the AAV inverted terminal repeat (ITRs) by electromobility shift assay. We present here several lines of evidence that transcription factors belonging to the RFX protein family bind specifically and selectively to AAV2 and AAV1 dsD sequences. Using supershift experiments, we characterize complexes containing RFX1 homodimers and RFX1/RFX3 heterodimers. Following transduction of HEK-293 cells, the AAV genome can be pulled-down by RFX1 and RFX3 antibodies. Moreover, our data suggest that RFX proteins which interact with transcriptional enhancers of several mammalian DNA viruses, can act as regulators of AAV mediated transgene expression.
Collapse
Affiliation(s)
- Laura Julien
- Sorbonne Universités UPMC Univ Paris 06, Inserm, Institut de Myologie, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, 105 bd de l'Hôpital, Paris, 13, France
| | - Julie Chassagne
- Sorbonne Universités UPMC Univ Paris 06, Inserm, Institut de Myologie, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, 105 bd de l'Hôpital, Paris, 13, France
| | - Cécile Peccate
- Sorbonne Universités UPMC Univ Paris 06, Inserm, Institut de Myologie, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, 105 bd de l'Hôpital, Paris, 13, France
| | - Stéphanie Lorain
- Sorbonne Universités UPMC Univ Paris 06, Inserm, Institut de Myologie, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, 105 bd de l'Hôpital, Paris, 13, France
| | - France Piétri-Rouxel
- Sorbonne Universités UPMC Univ Paris 06, Inserm, Institut de Myologie, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, 105 bd de l'Hôpital, Paris, 13, France
| | - Olivier Danos
- REGENXBIO, 9600 Blackwell Rd, Rockville, MD, 20850, USA
| | - Sofia Benkhelifa-Ziyyat
- Sorbonne Universités UPMC Univ Paris 06, Inserm, Institut de Myologie, Centre de Recherche en Myologie (CRM), GH Pitié Salpêtrière, 105 bd de l'Hôpital, Paris, 13, France.
| |
Collapse
|
219
|
Schön C, Becirovic E, Biel M, Michalakis S. Design and Development of AAV-based Gene Supplementation Therapies for Achromatopsia and Retinitis Pigmentosa. Methods Mol Biol 2018; 1715:33-46. [PMID: 29188504 DOI: 10.1007/978-1-4939-7522-8_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Achromatopsia (ACHM) and retinitis pigmentosa (RP) are inherited disorders caused by mutations in cone and rod photoreceptor-specific genes, respectively. ACHM strongly impairs daylight vision, whereas RP initially affects night vision and daylight vision at later stages. Currently, gene supplementation therapies utilizing recombinant adeno-associated virus (rAAV) vectors are being developed for various forms of ACHM and RP. In this chapter, we describe the procedure of designing and developing specific and efficient rAAV vectors for cone- and rod-specific gene supplementation.
Collapse
Affiliation(s)
- Christian Schön
- Department of Pharmacy, Center for Drug Research, Center for Integrated Protein Science Munich CiPSM, Ludwig-Maximilian-University, Munich, Germany
| | - Elvir Becirovic
- Department of Pharmacy, Center for Drug Research, Center for Integrated Protein Science Munich CiPSM, Ludwig-Maximilian-University, Munich, Germany
| | - Martin Biel
- Department of Pharmacy, Center for Drug Research, Center for Integrated Protein Science Munich CiPSM, Ludwig-Maximilian-University, Munich, Germany
| | - Stylianos Michalakis
- Department of Pharmacy, Center for Drug Research, Center for Integrated Protein Science Munich CiPSM, Ludwig-Maximilian-University, Munich, Germany.
| |
Collapse
|
220
|
Ahmed SG, Waddington SN, Boza-Morán MG, Yáñez-Muñoz RJ. High-efficiency transduction of spinal cord motor neurons by intrauterine delivery of integration-deficient lentiviral vectors. J Control Release 2017; 273:99-107. [PMID: 29289570 PMCID: PMC5845930 DOI: 10.1016/j.jconrel.2017.12.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Revised: 12/24/2017] [Accepted: 12/27/2017] [Indexed: 12/21/2022]
Abstract
Integration-deficient lentiviral vectors (IDLVs) are promising gene delivery tools that retain the high transduction efficiency of standard lentiviral vectors, yet fail to integrate as proviruses and are instead converted into episomal circles. These episomes are metabolically stable and support long-term expression of transgenes in non-dividing cells, exhibiting a decreased risk of insertional mutagenesis. We have embarked on an extensive study to compare the transduction efficiency of IDLVs pseudotyped with different envelopes (vesicular stomatitis, Rabies, Mokola and Ross River viral envelopes) and self-complementary adeno-associated viral vectors, serotype-9 (scAAV-9) in spinal cord tissues after intraspinal injection of mouse embryos (E16). Our results indicate that IDLVs can transduce motor neurons (MNs) at extremely high efficiency regardless of the envelope pseudotype while scAAV9 mediates gene delivery to ~ 40% of spinal cord motor neurons, with other non-neuronal cells also transduced. Long-term expression studies revealed stable gene expression at 7 months post-injection. Taken together, the results of this study indicate that IDLVs may be efficient tools for in utero cord transduction in therapeutic strategies such as for treatment of inherited early childhood neurodegenerative diseases.
Collapse
Affiliation(s)
- Sherif G Ahmed
- AGCTlab.org, Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway, University of London, Egham TW20 0EX, UK; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Egypt
| | - Simon N Waddington
- The Institute for Women's Health, University College London, London, UK; MRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Maria Gabriela Boza-Morán
- AGCTlab.org, Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway, University of London, Egham TW20 0EX, UK
| | - Rafael J Yáñez-Muñoz
- AGCTlab.org, Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway, University of London, Egham TW20 0EX, UK.
| |
Collapse
|
221
|
Han KS, Cooke SF, Xu W. Experience-Dependent Equilibration of AMPAR-Mediated Synaptic Transmission during the Critical Period. Cell Rep 2017; 18:892-904. [PMID: 28122240 DOI: 10.1016/j.celrep.2016.12.084] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 11/21/2016] [Accepted: 12/26/2016] [Indexed: 11/17/2022] Open
Abstract
Experience-dependent synapse refinement is essential for functional optimization of neural circuits. However, how sensory experience sculpts excitatory synaptic transmission is poorly understood. Here, we show that despite substantial remodeling of synaptic connectivity, AMPAR-mediated synaptic transmission remains at equilibrium during the critical period in the mouse primary visual cortex. The maintenance of this equilibrium requires neurogranin (Ng), a postsynaptic calmodulin-binding protein important for synaptic plasticity. With normal visual experience, loss of Ng decreased AMPAR-positive synapse numbers, prevented AMPAR-silent synapse maturation, and increased spine elimination. Importantly, visual deprivation halted synapse loss caused by loss of Ng, revealing that Ng coordinates experience-dependent AMPAR-silent synapse conversion to AMPAR-active synapses and synapse elimination. Loss of Ng also led to sensitized long-term synaptic depression (LTD) and impaired visually guided behavior. Our synaptic interrogation reveals that experience-dependent coordination of AMPAR-silent synapse conversion and synapse elimination hinges upon Ng-dependent mechanisms for constructive synaptic refinement during the critical period.
Collapse
Affiliation(s)
- Kyung-Seok Han
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Samuel F Cooke
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Weifeng Xu
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
222
|
Restoration of patterned vision with an engineered photoactivatable G protein-coupled receptor. Nat Commun 2017; 8:1862. [PMID: 29192252 PMCID: PMC5709376 DOI: 10.1038/s41467-017-01990-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 10/27/2017] [Indexed: 12/21/2022] Open
Abstract
Retinitis pigmentosa results in blindness due to degeneration of photoreceptors, but spares other retinal cells, leading to the hope that expression of light-activated signaling proteins in the surviving cells could restore vision. We used a retinal G protein-coupled receptor, mGluR2, which we chemically engineered to respond to light. In retinal ganglion cells (RGCs) of blind rd1 mice, photoswitch-charged mGluR2 (“SNAG-mGluR2”) evoked robust OFF responses to light, but not in wild-type retinas, revealing selectivity for RGCs that have lost photoreceptor input. SNAG-mGluR2 enabled animals to discriminate parallel from perpendicular lines and parallel lines at varying spacing. Simultaneous viral delivery of the inhibitory SNAG-mGluR2 and excitatory light-activated ionotropic glutamate receptor LiGluR yielded a distribution of expression ratios, restoration of ON, OFF and ON-OFF light responses and improved visual acuity. Thus, SNAG-mGluR2 restores patterned vision and combinatorial light response diversity provides a new logic for enhanced-acuity retinal prosthetics. To restore sight after retinal degeneration, one approach is to express light-sensitive proteins in remaining cells. Here the authors combine a light-sensitive engineered G protein-coupled receptor and ion channels to restore ON and OFF responses as well as superior visual pattern discrimination.
Collapse
|
223
|
Hartl D, Krebs AR, Jüttner J, Roska B, Schübeler D. Cis-regulatory landscapes of four cell types of the retina. Nucleic Acids Res 2017; 45:11607-11621. [PMID: 29059322 PMCID: PMC5714137 DOI: 10.1093/nar/gkx923] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/28/2017] [Accepted: 10/02/2017] [Indexed: 12/18/2022] Open
Abstract
The retina is composed of ∼50 cell-types with specific functions for the process of vision. Identification of the cis-regulatory elements active in retinal cell-types is key to elucidate the networks controlling this diversity. Here, we combined transcriptome and epigenome profiling to map the regulatory landscape of four cell-types isolated from mouse retinas including rod and cone photoreceptors as well as rare inter-neuron populations such as horizontal and starburst amacrine cells. Integration of this information reveals sequence determinants and candidate transcription factors for controlling cellular specialization. Additionally, we refined parallel reporter assays to enable studying the transcriptional activity of large collection of sequences in individual cell-types isolated from a tissue. We provide proof of concept for this approach and its scalability by characterizing the transcriptional capacity of several hundred putative regulatory sequences within individual retinal cell-types. This generates a catalogue of cis-regulatory regions active in retinal cell types and we further demonstrate their utility as potential resource for cellular tagging and manipulation.
Collapse
Affiliation(s)
- Dominik Hartl
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, CH 4058 Basel, Switzerland
- University of Basel, Faculty of Sciences, Petersplatz 1, CH 4003 Basel, Switzerland
| | - Arnaud R. Krebs
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, CH 4058 Basel, Switzerland
| | - Josephine Jüttner
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, CH 4058 Basel, Switzerland
| | - Botond Roska
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, CH 4058 Basel, Switzerland
- University of Basel, Department of Ophthalmology, Mittlere Strasse 91, CH 4031 Basel, Switzerland
| | - Dirk Schübeler
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, CH 4058 Basel, Switzerland
- University of Basel, Faculty of Sciences, Petersplatz 1, CH 4003 Basel, Switzerland
| |
Collapse
|
224
|
Liao H, Zhong Z, Liu Z, Li L, Ling Z, Zou X. Bone mesenchymal stem cells co-expressing VEGF and BMP-6 genes to combat avascular necrosis of the femoral head. Exp Ther Med 2017; 15:954-962. [PMID: 29399103 PMCID: PMC5772743 DOI: 10.3892/etm.2017.5455] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 11/03/2017] [Indexed: 02/06/2023] Open
Abstract
The aim of the present study was to investigate the potential of bone mesenchymal stem cells (BMSCs) treated with a combination of vascular endothelial growth factor (VEGF) and bone morphogenetic protein-6 (BMP-6) genes for the treatment of avascular necrosis of the femoral head (ANFH). Rat BMSCs were isolated and purified using a density gradient centrifugation method. The purity and characteristics of the BMSCs were detected by cell surface antigens identification using flow cytometry. The experimental groups were administered with one of the following adeno-associated virus (AAV) vector constructs: AAV-green fluorescent protein (AAV-GFP), AAV-BMP-6, AAV-VEGF or AAV-VEGF-BMP-6. The expression of VEGF and BMP-6 was detected by reverse transcription-quantitative polymerase chain reaction, western blotting and ELISA assays. The effects of VEGF and BMP-6 on BMSCs were evaluated by angiogenic and osteogenic assays. The transfected BMSCs were combined with a biomimetic synthetic scaffold poly lactide-co-glycolide (PLAGA) and they were then subcutaneously implanted into nude mice. After four weeks, the implants were analyzed with histology and subsequent immunostaining to evaluate the effects of BMSCs on blood vessel and bone formation in vivo. In the AAV-VEGF-BMP-6 group, the expression levels of VEGF and BMP-6 were significantly increased and human umbilical vein endothelial cells tube formation was significantly enhanced compared with other groups. Capillaries and bone formation in the AAV-VEGF-BMP-6 group was significantly higher compared with the other groups. The results of the present study suggest that BMSCs expressing both VEGF and BMP-6 induce an increase in blood vessels and bone formation, which provides theoretical support for ANFH gene therapy.
Collapse
Affiliation(s)
- Hongxing Liao
- Department of Orthopedics, Meizhou People's Hospital, Meizhou, Guangdong 514000, P.R. China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zhixiong Zhong
- Department of Cardiovascular Medicine, Meizhou People's Hospital, Meizhou, Guangdong 514000, P.R. China
| | - Zhanliang Liu
- Department of Orthopedics, Meizhou People's Hospital, Meizhou, Guangdong 514000, P.R. China
| | - Liangping Li
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zemin Ling
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
225
|
Gundry MC, Dever DP, Yudovich D, Bauer DE, Haas S, Wilkinson AC, Singbrant S. Technical considerations for the use of CRISPR/Cas9 in hematology research. Exp Hematol 2017; 54:4-11. [PMID: 28757433 PMCID: PMC5603407 DOI: 10.1016/j.exphem.2017.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 07/19/2017] [Indexed: 11/23/2022]
Abstract
The hematopoietic system is responsible for transporting oxygen and nutrients, fighting infections, and repairing tissue damage. Hematopoietic system dysfunction therefore causes a range of serious health consequences. Lifelong hematopoiesis is maintained by repopulating multipotent hematopoietic stem cells (HSCs) that replenish shorter-lived, mature blood cell types. A prokaryotic mechanism of immunity, the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 nuclease system, has been recently "repurposed" to mutate mammalian genomes efficiently and in a sequence-specific manner. The application of this genome-editing technology to hematology has afforded new approaches for functional genomics and even the prospect of "correcting" dysfunctional HSCs in the treatment of serious genetic hematological diseases. In this Perspective, we provide an overview of three recent CRISPR/Cas9 methods in hematology: gene disruption, gene targeting, and saturating mutagenesis. We also summarize the technical considerations and advice provided during the May 2017 International Society of Experimental Hematology New Investigator Committee webinar on the same topic.
Collapse
Affiliation(s)
- Michael C Gundry
- Department of Molecular and Human Genetics, Stem Cells and Regenerative Medicine Center, Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| | - Daniel P Dever
- Department of Pediatrics, Stanford University, Stanford, CA
| | - David Yudovich
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Daniel E Bauer
- Division of Hematology/Oncology, Boston Children's Hospital, Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Simon Haas
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine and Division of Stem Cells and Cancer, DKFZ German Cancer Research Centre, Heidelberg, Germany
| | - Adam C Wilkinson
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA.
| | - Sofie Singbrant
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
226
|
Chai Z, Sun J, Rigsbee KM, Wang M, Samulski RJ, Li C. Application of polyploid adeno-associated virus vectors for transduction enhancement and neutralizing antibody evasion. J Control Release 2017; 262:348-356. [PMID: 28789965 PMCID: PMC5819605 DOI: 10.1016/j.jconrel.2017.08.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 07/18/2017] [Accepted: 08/02/2017] [Indexed: 01/13/2023]
Abstract
Adeno-associated virus (AAV) vectors have been used successfully in clinical trials for patients with hemophilia or blindness, but pre-existing neutralizing antibodies (Nab) are common in the general population and exclude many patients from clinical trials. Exploration of effective strategies to enhance AAV transduction and escape from Nab activity is still imperative. Previous studies have shown the compatibility of capsids from AAV serotypes and homology of recognition sites of AAV Nab located on different capsid subunits from one virion. In this study, we co-transfected AAV2 and AAV8 helper plasmids at different ratios (3:1, 1:1 and 1:3) to assemble haploid capsids and study both their transduction efficiency and Nab escape activity. After muscular injection, all of the haploid viruses induced higher transduction than their parental AAV vectors (2- to 9-fold over AAV2), with the highest of these being the haploid vector AAV2/8 3:1. After systemic administration, a 4-fold higher transduction in the liver was observed with haploid AAV2/8 1:3 than that with AAV8 alone. We then packaged the therapeutic factor IX cassette into haploid AAV2/8 1:3 capsids and injected them into FIX knockout mice via the tail vein. Higher FIX expression and improved phenotypic correction were achieved with the haploid AAV2/8 1:3 virus vector when compared to that of AAV8. Additionally, the haploid virus AAV2/8 1:3 was able to escape AAV2 neutralization and did not increase capsid antigen presentation capacity when compared to AAV8. To improve the Nab evasion ability of the haploid virus, we produced the triploid vector AAV2/8/9 by co-transfecting AAV2, AAV8 and AAV9 helper plasmids at a ratio of 1:1:1. After systemic administration, a 2-fold higher transduction in the liver was observed with the triploid vector AAV2/8/9 than that with AAV8. Nab analysis demonstrated that the triploid AAV2/8/9 vector was able to escape Nab activity from mouse sera immunized with parental serotypes. These results indicate that polyploid viruses might potentially acquire advantages from parental serotypes for enhancement of AAV transduction and evasion of Nab recognition without increasing capsid antigen presentation in target cells. Polyploid AAV vectors can be generated from any AAV serotype, whether natural, rational, library derived or a combination thereof, providing a novel strategy that should be explored in future clinical trials in patients with neutralizing antibodies.
Collapse
Affiliation(s)
- Zheng Chai
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Junjiang Sun
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Kelly Michelle Rigsbee
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Mei Wang
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States; Institute of Hematology, Chinese Academy of Medical Sciences, Tianjin, China
| | - R Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States; Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States.
| |
Collapse
|
227
|
Jean-Charles PY, Yu SMW, Abraham D, Kommaddi RP, Mao L, Strachan RT, Zhang ZS, Bowles DE, Brian L, Stiber JA, Jones SN, Koch WJ, Rockman HA, Shenoy SK. Mdm2 regulates cardiac contractility by inhibiting GRK2-mediated desensitization of β-adrenergic receptor signaling. JCI Insight 2017; 2:95998. [PMID: 28878120 DOI: 10.1172/jci.insight.95998] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 07/27/2017] [Indexed: 12/22/2022] Open
Abstract
The oncoprotein Mdm2 is a RING domain-containing E3 ubiquitin ligase that ubiquitinates G protein-coupled receptor kinase 2 (GRK2) and β-arrestin2, thereby regulating β-adrenergic receptor (βAR) signaling and endocytosis. Previous studies showed that cardiac Mdm2 expression is critical for controlling p53-dependent apoptosis during early embryonic development, but the role of Mdm2 in the developed adult heart is unknown. We aimed to identify if Mdm2 affects βAR signaling and cardiac function in adult mice. Using Mdm2/p53-KO mice, which survive for 9-12 months, we identified a critical and potentially novel role for Mdm2 in the adult mouse heart through its regulation of cardiac β1AR signaling. While baseline cardiac function was mostly similar in both Mdm2/p53-KO and wild-type (WT) mice, isoproterenol-induced cardiac contractility in Mdm2/p53-KO was significantly blunted compared with WT mice. Isoproterenol increased cAMP in left ventricles of WT but not of Mdm2/p53-KO mice. Additionally, while basal and forskolin-induced calcium handling in isolated Mdm2/p53-KO and WT cardiomyocytes were equivalent, isoproterenol-induced calcium handling in Mdm2/p53-KO was impaired. Mdm2/p53-KO hearts expressed 2-fold more GRK2 than WT. GRK2 polyubiquitination via lysine-48 linkages was significantly reduced in Mdm2/p53-KO hearts. Tamoxifen-inducible cardiomyocyte-specific deletion of Mdm2 in adult mice also led to a significant increase in GRK2, and resulted in severely impaired cardiac function, high mortality, and no detectable βAR responsiveness. Gene delivery of either Mdm2 or GRK2-CT in vivo using adeno-associated virus 9 (AAV9) effectively rescued β1AR-induced cardiac contractility in Mdm2/p53-KO. These findings reveal a critical p53-independent physiological role of Mdm2 in adult hearts, namely, regulation of GRK2-mediated desensitization of βAR signaling.
Collapse
Affiliation(s)
| | | | | | | | - Lan Mao
- Department of Medicine, Division of Cardiology, and
| | | | | | - Dawn E Bowles
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Leigh Brian
- Department of Medicine, Division of Cardiology, and
| | | | - Stephen N Jones
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Walter J Koch
- Center for Translational Medicine, Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Howard A Rockman
- Department of Medicine, Division of Cardiology, and.,Department of Cell Biology, and.,Department of Molecular Genetics, Duke University Medical Center, Durham, North Carolina, USA
| | - Sudha K Shenoy
- Department of Medicine, Division of Cardiology, and.,Department of Cell Biology, and
| |
Collapse
|
228
|
Huang X, Zhou G, Wu W, Duan Y, Ma G, Song J, Xiao R, Vandenberghe L, Zhang F, D'Amore PA, Lei H. Genome editing abrogates angiogenesis in vivo. Nat Commun 2017; 8:112. [PMID: 28740073 PMCID: PMC5524639 DOI: 10.1038/s41467-017-00140-3] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 06/05/2017] [Indexed: 12/29/2022] Open
Abstract
Angiogenesis, in which vascular endothelial growth factor receptor (VEGFR) 2 plays an essential role, is associated with a variety of human diseases including proliferative diabetic retinopathy and wet age-related macular degeneration. Here we report that a system of adeno-associated virus (AAV)-mediated clustered regularly interspaced short palindromic repeats (CRISPR)-associated endonuclease (Cas)9 from Streptococcus pyogenes (SpCas9) is used to deplete VEGFR2 in vascular endothelial cells (ECs), whereby the expression of SpCas9 is driven by an endothelial-specific promoter of intercellular adhesion molecule 2. We further show that recombinant AAV serotype 1 (rAAV1) transduces ECs of pathologic vessels, and that editing of genomic VEGFR2 locus using rAAV1-mediated CRISPR/Cas9 abrogates angiogenesis in the mouse models of oxygen-induced retinopathy and laser-induced choroid neovascularization. This work establishes a strong foundation for genome editing as a strategy to treat angiogenesis-associated diseases. Abnormal angiogenesis causes many ocular diseases. Here the authors employ CRISPR/Cas9 gene editing technology to silence VEGFR2, a major regulator of angiogenesis, in retinal endothelium and abrogate angiogenesis in the mouse models of oxygen-induced retinopathy and laser-induced choroid neovascularization.
Collapse
Affiliation(s)
- Xionggao Huang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.,Hainan Eye Hospital, Haikou, Hainan Province, 570311, China
| | - Guohong Zhou
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.,Shanxi Eye Hospital, Taiyuan, Shanxi Province, 030002, China
| | - Wenyi Wu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.,Department of Ophthalmology, Second Xiangya Hospital, Second Xiangya Hospital, Central South University, Changsha, Hunan Province, 410013, China
| | - Yajian Duan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.,Shanxi Eye Hospital, Taiyuan, Shanxi Province, 030002, China
| | - Gaoen Ma
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Jingyuan Song
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.,Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
| | - Ru Xiao
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Luk Vandenberghe
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Feng Zhang
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, 02142, USA
| | - Patricia A D'Amore
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA
| | - Hetian Lei
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA. .,Department of Ophthalmology, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
229
|
Deletion of the B-B' and C-C' regions of inverted terminal repeats reduces rAAV productivity but increases transgene expression. Sci Rep 2017; 7:5432. [PMID: 28710345 PMCID: PMC5511163 DOI: 10.1038/s41598-017-04054-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 05/09/2017] [Indexed: 11/30/2022] Open
Abstract
Inverted terminal repeats (ITRs) of the adeno-associated virus (AAV) are essential for rescue, replication, packaging, and integration of the viral genome. While ITR mutations have been identified in previous reports, we designed a new truncated ITR lacking the B-B’ and C-C’ regions named as ITRΔBC and investigated its effects on viral genome replication, packaging, and expression of recombinant AAV (rAAV). The packaging ability was compared between ITRΔBC rAAV and wild-type (wt) ITR rAAV. Our results showed the productivity of ITRΔBC rAAV was reduced 4-fold, which is consistent with the 8-fold decrease in the replication of viral genomic DNA of ITRΔBC rAAV compared with wt ITR rAAV. Surprisingly, transgene expression was significantly higher for ITRΔBC rAAV. A preliminary exploration of the underlying mechanisms was carried out by inhibiting and degrading the ataxia telangiectasia mutated (ATM) protein and the Mre11 complex (MRN), respectively, since the rAAV expression was inhibited by the ATM and/or MRN through cis interaction or binding with wt ITRs. We demonstrated that the inhibitory effects were weakened on ITRΔBC rAAV expression. This study suggests deletion in ITR can affect the transgene expression of AAV, which provides a new way to improve the AAV expression through ITRs modification.
Collapse
|
230
|
Cell Cycle-Dependent Expression of Adeno-Associated Virus 2 (AAV2) Rep in Coinfections with Herpes Simplex Virus 1 (HSV-1) Gives Rise to a Mosaic of Cells Replicating either AAV2 or HSV-1. J Virol 2017; 91:JVI.00357-17. [PMID: 28515305 DOI: 10.1128/jvi.00357-17] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 05/05/2017] [Indexed: 12/19/2022] Open
Abstract
Adeno-associated virus 2 (AAV2) depends on the simultaneous presence of a helper virus such as herpes simplex virus 1 (HSV-1) for productive replication. At the same time, AAV2 efficiently blocks the replication of HSV-1, which would eventually limit its own replication by diminishing the helper virus reservoir. This discrepancy begs the question of how AAV2 and HSV-1 can coexist in a cell population. Here we show that in coinfected cultures, AAV2 DNA replication takes place almost exclusively in S/G2-phase cells, while HSV-1 DNA replication is restricted to G1 phase. Live microscopy revealed that not only wild-type AAV2 (wtAAV2) replication but also reporter gene expression from both single-stranded and double-stranded (self-complementary) recombinant AAV2 vectors preferentially occurs in S/G2-phase cells, suggesting that the preference for S/G2 phase is independent of the nature of the viral genome. Interestingly, however, a substantial proportion of S/G2-phase cells transduced by the double-stranded but not the single-stranded recombinant AAV2 vectors progressed through mitosis in the absence of the helper virus. We conclude that cell cycle-dependent AAV2 rep expression facilitates cell cycle-dependent AAV2 DNA replication and inhibits HSV-1 DNA replication. This may limit competition for cellular and viral helper factors and, hence, creates a biological niche for either virus to replicate.IMPORTANCE Adeno-associated virus 2 (AAV2) differs from most other viruses, as it requires not only a host cell for replication but also a helper virus such as an adenovirus or a herpesvirus. This situation inevitably leads to competition for cellular resources. AAV2 has been shown to efficiently inhibit the replication of helper viruses. Here we present a new facet of the interaction between AAV2 and one of its helper viruses, herpes simplex virus 1 (HSV-1). We observed that AAV2 rep gene expression is cell cycle dependent and gives rise to distinct time-controlled windows for HSV-1 replication. High Rep protein levels in S/G2 phase support AAV2 replication and inhibit HSV-1 replication. Conversely, low Rep protein levels in G1 phase permit HSV-1 replication but are insufficient for AAV2 replication. This allows both viruses to productively replicate in distinct sets of dividing cells.
Collapse
|
231
|
Fenno LE, Mattis J, Ramakrishnan C, Deisseroth K. A Guide to Creating and Testing New INTRSECT Constructs. ACTA ACUST UNITED AC 2017; 80:4.39.1-4.39.24. [PMID: 28678399 PMCID: PMC6892587 DOI: 10.1002/cpns.30] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
As the power of genetically encoded interventional and observational tools for neuroscience expands, the boundaries of experimental design are increasingly defined by limits in selectively expressing these tools in relevant cell types. Single-recombinase-dependent expression systems have been widely used as a means to restrict gene expression based on single features by combining recombinase-dependent viruses with recombinase-expressing transgenic animals. This protocol details how to create INTRSECT constructs and use multiple recombinases to achieve targeting of a desired gene to subsets of neurons that are defined by multiple genetic and/or topological features. This method includes the design and utilization of both viruses and transgenic animals: these tools are inherently flexible and modular and may be used in different combinations to achieve the desired gene expression pattern. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Lief E Fenno
- Department of Bioengineering, Stanford University, Stanford, California.,Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Joanna Mattis
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, California.,Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California.,CNC Program, Stanford University, Stanford, California.,Howard Hughes Medical Institute, Stanford University, Stanford, California
| |
Collapse
|
232
|
Bernier BE, Lacagnina AF, Ayoub A, Shue F, Zemelman BV, Krasne FB, Drew MR. Dentate Gyrus Contributes to Retrieval as well as Encoding: Evidence from Context Fear Conditioning, Recall, and Extinction. J Neurosci 2017; 37:6359-6371. [PMID: 28546308 PMCID: PMC5490069 DOI: 10.1523/jneurosci.3029-16.2017] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 04/10/2017] [Accepted: 05/20/2017] [Indexed: 11/21/2022] Open
Abstract
Dentate gyrus (DG) is widely thought to provide a teaching signal that enables hippocampal encoding of memories, but its role during retrieval is poorly understood. Some data and models suggest that DG plays no role in retrieval; others encourage the opposite conclusion. To resolve this controversy, we evaluated the effects of optogenetic inhibition of dorsal DG during context fear conditioning, recall, generalization, and extinction in male mice. We found that (1) inhibition during training impaired context fear acquisition; (2) inhibition during recall did not impair fear expression in the training context, unless mice had to distinguish between similar feared and neutral contexts; (3) inhibition increased generalization of fear to an unfamiliar context that was similar to a feared one and impaired fear expression in the conditioned context when it was similar to a neutral one; and (4) inhibition impaired fear extinction. These effects, as well as several seemingly contradictory published findings, could be reproduced by BACON (Bayesian Context Fear Algorithm), a physiologically realistic hippocampal model positing that acquisition and retrieval both involve coordinated activity in DG and CA3. Our findings thus suggest that DG contributes to retrieval and extinction, as well as to the initial establishment of context fear.SIGNIFICANCE STATEMENT Despite abundant evidence that the hippocampal dentate gyrus (DG) plays a critical role in memory, it remains unclear whether the role of DG relates to memory acquisition or retrieval. Using contextual fear conditioning and optogenetic inhibition, we show that DG contributes to both of these processes. Using computational simulations, we identify specific mechanisms through which the suppression of DG affects memory performance. Finally, we show that DG contributes to fear extinction learning, a process in which learned fear is attenuated through exposures to a fearful context in the absence of threat. Our data resolve a long-standing question about the role of DG in memory and provide insight into how disorders affecting DG, including aging, stress, and depression, influence cognitive processes.
Collapse
Affiliation(s)
- Brian E Bernier
- Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin, Texas 78712, and
| | - Anthony F Lacagnina
- Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin, Texas 78712, and
| | - Adam Ayoub
- Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin, Texas 78712, and
| | - Francis Shue
- Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin, Texas 78712, and
| | - Boris V Zemelman
- Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin, Texas 78712, and
| | - Franklin B Krasne
- Department of Psychology, University of California at Los Angeles, Los Angeles, California 90095
| | - Michael R Drew
- Center for Learning and Memory, Department of Neuroscience, University of Texas at Austin, Austin, Texas 78712, and
| |
Collapse
|
233
|
Sun Y, Liu CH, Wang Z, Meng SS, Burnim SB, SanGiovanni JP, Kamenecka TM, Solt LA, Chen J. RORα modulates semaphorin 3E transcription and neurovascular interaction in pathological retinal angiogenesis. FASEB J 2017. [PMID: 28646017 DOI: 10.1096/fj.201700172r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Pathological proliferation of retinal blood vessels commonly causes vision impairment in proliferative retinopathies, including retinopathy of prematurity. Dysregulated crosstalk between the vasculature and retinal neurons is increasingly recognized as a major factor contributing to the pathogenesis of vascular diseases. Class 3 semaphorins (SEMA3s), a group of neuron-secreted axonal and vascular guidance factors, suppress pathological vascular growth in retinopathy. However, the upstream transcriptional regulators that mediate the function of SEMA3s in vascular growth are poorly understood. Here we showed that retinoic acid receptor-related orphan receptor α (RORα), a nuclear receptor and transcription factor, is a novel transcriptional regulator of SEMA3E-mediated neurovascular coupling in a mouse model of oxygen-induced proliferative retinopathy. We found that genetic deficiency of RORα substantially induced Sema3e expression in retinopathy. Both RORα and SEMA3E were expressed in retinal ganglion cells. RORα directly bound to a specific ROR response element on the promoter of Sema3e and negatively regulated Sema3e promoter-driven luciferase expression. Suppression of Sema3e using adeno-associated virus 2 carrying short hairpin RNA targeting Sema3e promoted disoriented pathological neovascularization and partially abolished the inhibitory vascular effects of RORα deficiency in retinopathy. Our findings suggest that RORα is a novel transcriptional regulator of SEMA3E-mediated neurovascular coupling in pathological retinal angiogenesis.-Sun, Y., Liu, C.-H., Wang, Z., Meng, S. S., Burnim, S. B., SanGiovanni, J. P., Kamenecka, T. M., Solt, L. A., Chen, J. RORα modulates semaphorin 3E transcription and neurovascular interaction in pathological retinal angiogenesis.
Collapse
Affiliation(s)
- Ye Sun
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Chi-Hsiu Liu
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Zhongxiao Wang
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Steven S Meng
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Samuel B Burnim
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - John Paul SanGiovanni
- Section of Nutritional Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA.,Department of Biochemistry and Molecular and Cellular Biology, Georgetown School of Medicine, Washington, D.C., USA
| | - Theodore M Kamenecka
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida, USA
| | - Laura A Solt
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Jing Chen
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA;
| |
Collapse
|
234
|
Abstract
Dysferlinopathy is an autosomal recessive muscular dystrophy characterized by the progressive loss of motility that is caused by mutations throughout the DYSF gene. There are currently no approved therapies that ameliorate or reverse dysferlinopathy. Gene delivery using adeno-associated vectors (AAVs) is a leading therapeutic strategy for genetic diseases; however, the large size of dysferlin cDNA (6.2 kB) precludes packaging into a single AAV capsid. Therefore, using 3D structural modeling and hypothesizing dysferlin C2 domain redundancy, a 30% smaller, dysferlin-like molecule amenable to single AAV vector packaging was engineered (termed Nano-Dysferlin). The intracellular distribution of Nano-Dysferlin was similar to wild-type dysferlin and neither demonstrated toxicity when overexpressed in dysferlin-deficient patient myoblasts. Intramuscular injection of AAV-Nano-Dysferlin in young dysferlin-deficient mice significantly improved muscle integrity and decreased muscle turnover 3 weeks after treatment, as determined by Evans blue dye uptake and central nucleated fibers, respectively. Systemically administered AAV-Nano-Dysferlin to young adult dysferlin-deficient mice restored motor function and improved muscle integrity nearly 8 months after a single injection. These preclinical data are the first report of a smaller dysferlin variant tailored for AAV single particle delivery that restores motor function and, therefore, represents an attractive candidate for the treatment of dysferlinopathy.
Collapse
|
235
|
Liang KJ, Woodard KT, Weaver MA, Gaylor JP, Weiss ER, Samulski RJ. AAV-Nrf2 Promotes Protection and Recovery in Animal Models of Oxidative Stress. Mol Ther 2017; 25:765-779. [PMID: 28253482 DOI: 10.1016/j.ymthe.2016.12.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 11/07/2016] [Accepted: 12/14/2016] [Indexed: 12/24/2022] Open
Abstract
NRF2 is a transcription factor that drives antioxidant gene expression in multiple organ systems. We hypothesized that Nrf2 overexpression could be therapeutically applied toward diseases in which redox homeostasis is disrupted. In this study, adeno-associated virus (AAV)-Nrf2 was tested in a mouse model of acute acetaminophen-induced liver toxicity and successfully conferred protection from hepatotoxicity, validating the vector design and early onset of NRF2-mediated protection. Furthermore, therapeutic potential of AAV-Nrf2 in chronic disease also was tested in a light-induced mouse model of age-related macular degeneration. Adult BALB/c mice were intravitreally injected with AAV-Nrf2 and subject to light damage following injection. Retinal thickness and function were monitored following light damage using optical coherence tomography and electroretinography, respectively. By 3 months post-damage, injected eyes had greater retinal thickness compared to uninjected controls. At 1 month post-damage, AAV-Nrf2 injection facilitated full functional recovery from light damage. Our results suggest a therapeutic potential for Nrf2 overexpression in acute and long-term capacities in multiple organ systems, opening up doors for combination gene therapy where replacement gene therapy requires additional therapeutic support to prevent further degeneration.
Collapse
Affiliation(s)
- Katharine J Liang
- Gene Therapy Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kenton T Woodard
- Gene Therapy Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mark A Weaver
- Departments of Medicine and Biostatistics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - John Paul Gaylor
- Gene Therapy Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ellen R Weiss
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - R Jude Samulski
- Gene Therapy Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
236
|
Sanchez AC, Li C, Andrews B, Asenjo JA, Samulski RJ. AAV Gene Therapy for Alcoholism: Inhibition of Mitochondrial Aldehyde Dehydrogenase Enzyme Expression in Hepatoma Cells. Hum Gene Ther 2017; 28:717-725. [PMID: 28578603 DOI: 10.1089/hum.2017.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Most ethanol is broken down in the liver in two steps by alcohol dehydrogenase (ADH) and aldehyde dehydrogenase (ALDH2) enzymes, which metabolize down ethanol into acetaldehyde and then acetate. Some individuals from the Asian population who carry a mutation in the aldehyde dehydrogenase gene (ALDH2*2) cannot metabolize acetaldehyde as efficiently, producing strong effects, including facial flushing, dizziness, hypotension, and palpitations. This results in an aversion to alcohol intake and protection against alcoholism. The large prevalence of this mutation in the human population strongly suggests that modulation of ALDH2 expression by genetic technologies could result in a similar phenotype. scAAV2 vectors encoding ALDH2 small hairpin RNA (shRNA) were utilized to validate this hypothesis by silencing ALDH2 gene expression in human cell lines. Human cell lines HEK-293 and HepG2 were transduced with scAAV2/shRNA, showing a reduction in ALDH2 RNA and protein expression with the two viral concentration assayed (1 × 104 and 1 × 105 vg/cell) at two different time points. In both cell lines, ALDH2 RNA levels were reduced by 90% and protein expression was inhibited by 90% and 52%, respectively, 5 days post infection. Transduced HepG2 VL17A cells (ADH+) exposed to ethanol resulted in a 50% increase in acetaldehyde levels. These results suggest that gene therapy could be a useful tool for the treatment of alcoholism by knocking down ALDH2 expression using shRNA technology delivered by AAV vectors.
Collapse
Affiliation(s)
- Anamaria C Sanchez
- 1 Centre for Biotechnology and Bioengineering (CeBiB), Department of Chemical Engineering and Biotechnology, University of Chile , Santiago, Chile
| | - Chengwen Li
- 2 Gene Therapy Center, University of North Carolina , Chapel Hill, North Carolina
| | - Barbara Andrews
- 1 Centre for Biotechnology and Bioengineering (CeBiB), Department of Chemical Engineering and Biotechnology, University of Chile , Santiago, Chile
| | - Juan A Asenjo
- 1 Centre for Biotechnology and Bioengineering (CeBiB), Department of Chemical Engineering and Biotechnology, University of Chile , Santiago, Chile
| | - R Jude Samulski
- 2 Gene Therapy Center, University of North Carolina , Chapel Hill, North Carolina
| |
Collapse
|
237
|
Hillier D, Fiscella M, Drinnenberg A, Trenholm S, Rompani SB, Raics Z, Katona G, Juettner J, Hierlemann A, Rozsa B, Roska B. Causal evidence for retina-dependent and -independent visual motion computations in mouse cortex. Nat Neurosci 2017; 20:960-968. [PMID: 28530661 PMCID: PMC5490790 DOI: 10.1038/nn.4566] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 04/22/2017] [Indexed: 12/14/2022]
Abstract
How neuronal computations in the sensory periphery contribute to computations in the cortex is not well understood. We examined this question in the context of visual-motion processing in the retina and primary visual cortex (V1) of mice. We disrupted retinal direction selectivity, either exclusively along the horizontal axis using FRMD7 mutants or along all directions by ablating starburst amacrine cells, and monitored neuronal activity in layer 2/3 of V1 during stimulation with visual motion. In control mice, we found an over-representation of cortical cells preferring posterior visual motion, the dominant motion direction an animal experiences when it moves forward. In mice with disrupted retinal direction selectivity, the over-representation of posterior-motion-preferring cortical cells disappeared, and their responses at higher stimulus speeds were reduced. This work reveals the existence of two functionally distinct, sensory-periphery-dependent and -independent computations of visual motion in the cortex.
Collapse
Affiliation(s)
- Daniel Hillier
- Neural Circuits Laboratory, Friedrich Miescher Institute, Basel, Switzerland
| | - Michele Fiscella
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Antonia Drinnenberg
- Neural Circuits Laboratory, Friedrich Miescher Institute, Basel, Switzerland
| | - Stuart Trenholm
- Neural Circuits Laboratory, Friedrich Miescher Institute, Basel, Switzerland
| | - Santiago B Rompani
- Neural Circuits Laboratory, Friedrich Miescher Institute, Basel, Switzerland
| | - Zoltan Raics
- Neural Circuits Laboratory, Friedrich Miescher Institute, Basel, Switzerland
| | - Gergely Katona
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.,The Faculty of Information Technology, Pázmány Péter Catholic University, Budapest, Hungary
| | - Josephine Juettner
- Neural Circuits Laboratory, Friedrich Miescher Institute, Basel, Switzerland
| | - Andreas Hierlemann
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Balazs Rozsa
- Laboratory of 3D Functional Network and Dendritic Imaging, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Botond Roska
- Neural Circuits Laboratory, Friedrich Miescher Institute, Basel, Switzerland.,Department of Ophthalmology, University of Basel, Basel, Switzerland
| |
Collapse
|
238
|
Heller KN, Mendell JT, Mendell JR, Rodino-Klapac LR. MicroRNA-29 overexpression by adeno-associated virus suppresses fibrosis and restores muscle function in combination with micro-dystrophin. JCI Insight 2017; 2:93309. [PMID: 28469083 DOI: 10.1172/jci.insight.93309] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 04/04/2017] [Indexed: 12/30/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by dystrophin deficiency resulting in progressive muscle weakness and fibrotic scarring. Muscle fibrosis impairs blood flow, hampering muscle repair and regeneration. Irrespective of the success of gene restoration, functional improvement is limited without reducing fibrosis. The levels of miR-29c, a known regulator of collagen, are reduced in DMD. Our goal is to develop translational, antifibrotic therapy by overexpressing miR-29c. We injected the gastrocnemius muscle with either self-complementary AAV.CMV.miR-29c or single-stranded AAV.MCK.micro-dystrophin alone or in combination in the mdx/utrn+/- mouse, a DMD mouse model. Treatment of 3-month-old mdx/utrn+/- mice with AAV.miR-29c showed a reduction in collagen and increased absolute and specific force compared with untreated animals, but neither parameter reached WT levels. Combinatorial gene delivery in 3-month-old mdx/utrn+/- mice further decreased fibrosis, and showed a reduction of transcript levels for Col1A, Col3A, fibronectin, and Tgfb1. In addition, absolute and specific force was normalized and equivalent to WT. However, protection against eccentric contraction fell short of WT levels at this time point. When this same mouse model was treated with miR-29c/micro-dystrophin combinatorial therapy at 1 month of age, there was complete normalization of specific and absolute force and protection against eccentric contraction-induced injury was comparable to WT. These findings highlight the potential for miR-29c as an important addition to the armamentarium for translational gene therapy, especially when used in combination with micro-dystrophin in DMD.
Collapse
Affiliation(s)
- Kristin N Heller
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics and Neurology, The Ohio State University, Columbus, Ohio, USA
| | - Joshua T Mendell
- Department of Molecular Biology.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jerry R Mendell
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics and Neurology, The Ohio State University, Columbus, Ohio, USA
| | - Louise R Rodino-Klapac
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics and Neurology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
239
|
Sun Y, Lin Z, Liu CH, Gong Y, Liegl R, Fredrick TW, Meng SS, Burnim SB, Wang Z, Akula JD, Pu WT, Chen J, Smith LEH. Inflammatory signals from photoreceptor modulate pathological retinal angiogenesis via c-Fos. J Exp Med 2017; 214:1753-1767. [PMID: 28465464 PMCID: PMC5461000 DOI: 10.1084/jem.20161645] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 02/02/2017] [Accepted: 03/22/2017] [Indexed: 11/04/2022] Open
Abstract
Pathological neovessels growing into the normally avascular photoreceptors cause vision loss in many eye diseases, such as age-related macular degeneration and macular telangiectasia. Ocular neovascularization is strongly associated with inflammation, but the source of inflammatory signals and the mechanisms by which these signals regulate the disruption of avascular privilege in photoreceptors are unknown. In this study, we found that c-Fos, a master inflammatory regulator, was increased in photoreceptors in a model of pathological blood vessels invading photoreceptors: the very low-density lipoprotein receptor-deficient (Vldlr-/- ) mouse. Increased c-Fos induced inflammatory cytokines interleukin 6 (IL-6) and tumor necrosis factor (TNF), leading to activation of signal transducer and activator of transcription 3 (STAT3) and increased TNFα-induced protein 3 (TNFAIP3) in Vldlr-/- photoreceptors. IL-6 activated the STAT3/vascular endothelial growth factor A (VEGFA) pathway directly, and elevated TNFAIP3 suppressed SOCS3 (suppressor of cytokine signaling 3)-activated STAT3/VEGFA indirectly. Inhibition of c-Fos using photoreceptor-specific AAV (adeno-associated virus)-hRK (human rhodopsin kinase)-sh_c-fos or a chemical inhibitor substantially reduced the pathological neovascularization and rescued visual function in Vldlr-/- mice. These findings suggested that the photoreceptor c-Fos controls blood vessel growth into the normally avascular photoreceptor layer through the inflammatory signal-induced STAT3/VEGFA pathway.
Collapse
Affiliation(s)
- Ye Sun
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - Zhiqiang Lin
- Department of Cardiology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - Chi-Hsiu Liu
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - Yan Gong
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - Raffael Liegl
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - Thomas W Fredrick
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - Steven S Meng
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - Samuel B Burnim
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - Zhongxiao Wang
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - James D Akula
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - William T Pu
- Department of Cardiology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| | - Jing Chen
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - Lois E H Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| |
Collapse
|
240
|
Rolland AS, Kareva T, Kholodilov N, Burke RE. A quantitative evaluation of a 2.5-kb rat tyrosine hydroxylase promoter to target expression in ventral mesencephalic dopamine neurons in vivo. Neuroscience 2017; 346:126-134. [PMID: 28108256 DOI: 10.1016/j.neuroscience.2017.01.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/15/2016] [Accepted: 01/09/2017] [Indexed: 01/08/2023]
Abstract
Adeno-associated viruses (AAVs) have become powerful tools in neuroscience for both basic research and potential therapeutic use. They have become especially important tools for optogenetic experiments based on their ability to achieve transgene expression in postmitotic neurons with regional selectivity. With the use of appropriate promoter elements they can achieve cellular specificity as well. One population of neurons that plays a central role in human neurodegenerative and psychiatric diseases are the dopamine neurons of the midbrain. To study these neurons in vivo with advanced techniques it would be highly advantageous to characterize an appropriate specific promoter. To this end we have characterized a 2.5-kb sequence of the rat tyrosine hydroxylase (TH) promoter. The rTHp(2.5) promoter induced expression of the fluorescent reporter protein mCherry in SN dopamine neurons. Although it showed excellent specificity in cortex and striatum, where no reporter expression was observed, in the SN region many neurons expressed reporter but not TH. We show that some of the TH negativity is due to the suppression of its expression by the transgene. We conclude that rTHp(2.5) does preferentially label dopamine neurons but its specificity is not complete within the substantia nigra and caution must be used.
Collapse
Affiliation(s)
- Anne-Sophie Rolland
- Department of Neurology, Columbia University, 650 W 168th Street, New York, NY 10032, USA
| | - Tatyana Kareva
- Department of Neurology, Columbia University, 650 W 168th Street, New York, NY 10032, USA
| | - Nikolai Kholodilov
- Department of Neurology, Columbia University, 650 W 168th Street, New York, NY 10032, USA
| | - Robert E Burke
- Department of Neurology, Columbia University, 650 W 168th Street, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University, 650 W 168th Street, New York, NY 10032, USA.
| |
Collapse
|
241
|
Sun Y, Liegl R, Gong Y, Bühler A, Cakir B, Meng SS, Burnim SB, Liu CH, Reuer T, Zhang P, Walz JM, Ludwig F, Lange C, Agostini H, Böhringer D, Schlunck G, Smith LEH, Stahl A. Sema3f Protects Against Subretinal Neovascularization In Vivo. EBioMedicine 2017; 18:281-287. [PMID: 28373097 PMCID: PMC5405173 DOI: 10.1016/j.ebiom.2017.03.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/17/2017] [Accepted: 03/17/2017] [Indexed: 12/26/2022] Open
Abstract
Pathological neovascularization of the outer retina is the hallmark of neovascular age-related macular degeneration (nAMD). Building on our previous observations that semaphorin 3F (Sema3f) is expressed in the outer retina and demonstrates anti-angiogenic potential, we have investigated whether Sema3f can be used to protect against subretinal neovascularization in two mouse models. Both in the very low-density lipid-receptor knockout (Vldlr−/−) model of spontaneous subretinal neovascularization as well as in the mouse model of laser-induced choroidal neovascularization (CNV), we found protective effects of Sema3f against the formation of pathologic neovascularization. In the Vldlr−/− model, AAV-induced overexpression of Sema3f reduced the size of pathologic neovascularization by 56%. In the laser-induced CNV model, intravitreally injected Sema3f reduced pathologic neovascularization by 30%. Combined, these results provide the first evidence from two distinct in vivo models for a use of Sema3f in protecting the outer retina against subretinal neovascularization. Sema3f is expressed in the physiologically avascular layers of the outer retina. Vldlr−/− mice have reduced Sema3f and form spontaneous subretinal neovascularization. AAV-mediated increase of Sema3f protects against neovascularization in Vldlr−/− mice. Sema3f also reduces pathologic neovascularization in eyes with laser-induced CNV.
Abnormal formation of new blood vessels in the retina is one of the hallmarks of a potentially blinding eye disease called wet (or exudative) macular degeneration. Here we investigated in two independent mouse models whether Sema3f (a protein involved in guiding blood vessel growth) can be modulated to protect against abnormal blood vessel growth. In both mouse models, we found protective effects of Sema3f against abnormal blood vessel formation in the retina. Combined, these results provide the first evidence that Sema3f could be modulated to protect against wet macular degeneration.
Collapse
Affiliation(s)
- Ye Sun
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Raffael Liegl
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Yan Gong
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Anima Bühler
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Bertan Cakir
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Steven S Meng
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Samuel B Burnim
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Chi-Hsiu Liu
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Tristan Reuer
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Peipei Zhang
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Johanna M Walz
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Franziska Ludwig
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Clemens Lange
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Hansjürgen Agostini
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Daniel Böhringer
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Günther Schlunck
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany
| | - Lois E H Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Andreas Stahl
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Killianstrasse 5, 79106 Freiburg, Germany.
| |
Collapse
|
242
|
Pozsgai ER, Griffin DA, Heller KN, Mendell JR, Rodino-Klapac LR. Systemic AAV-Mediated β-Sarcoglycan Delivery Targeting Cardiac and Skeletal Muscle Ameliorates Histological and Functional Deficits in LGMD2E Mice. Mol Ther 2017; 25:855-869. [PMID: 28284983 DOI: 10.1016/j.ymthe.2017.02.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 02/09/2017] [Accepted: 02/10/2017] [Indexed: 01/22/2023] Open
Abstract
Limb-girdle muscular dystrophy type 2E (LGMD2E), resulting from mutations in β-sarcoglycan (SGCB), is a progressive dystrophy with deteriorating muscle function, respiratory failure, and cardiomyopathy in 50% or more of LGMD2E patients. SGCB knockout mice share many of the phenotypic deficiencies of LGMD2E patients. To investigate systemic SGCB gene transfer to treat skeletal and cardiac muscle deficits, we designed a self-complementary AAVrh74 vector containing a codon-optimized human SGCB transgene driven by a muscle-specific promoter. We delivered scAAV.MHCK7.hSGCB through the tail vein of SGCB-/- mice to provide a rationale for a clinical trial that would lead to clinically meaningful results. This led to 98.1% transgene expression across all muscles that was accompanied by improvements in histopathology. Serum creatine kinase (CK) levels were reduced following treatment by 85.5%. Diaphragm force production increased by 94.4%, kyphoscoliosis of the spine was significantly reduced by 48.1%, overall ambulation increased by 57%, and vertical rearing increased dramatically by 132% following treatment. Importantly, no adverse effects were seen in muscle of wild-type mice injected systemically with scAAV.hSGCB. In this well-defined model of LGMD2E, we have demonstrated the efficacy and safety of systemic scAAV.hSGCB delivery, and these findings have established a path for clinically beneficial AAV-mediated gene therapy for LGMD2E.
Collapse
Affiliation(s)
- Eric R Pozsgai
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA; Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Danielle A Griffin
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Kristin N Heller
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Jerry R Mendell
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA; Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA; Department of Pediatrics and Neurology, The Ohio State University, Columbus, OH 43210, USA
| | - Louise R Rodino-Klapac
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210, USA; Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA; Department of Pediatrics and Neurology, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
243
|
Integration of a CD19 CAR into the TCR Alpha Chain Locus Streamlines Production of Allogeneic Gene-Edited CAR T Cells. Mol Ther 2017; 25:949-961. [PMID: 28237835 PMCID: PMC5383629 DOI: 10.1016/j.ymthe.2017.02.005] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 02/05/2017] [Accepted: 02/06/2017] [Indexed: 11/25/2022] Open
Abstract
Adoptive cellular therapy using chimeric antigen receptor (CAR) T cell therapies have produced significant objective responses in patients with CD19+ hematological malignancies, including durable complete responses. Although the majority of clinical trials to date have used autologous patient cells as the starting material to generate CAR T cells, this strategy poses significant manufacturing challenges and, for some patients, may not be feasible because of their advanced disease state or difficulty with manufacturing suitable numbers of CAR T cells. Alternatively, T cells from a healthy donor can be used to produce an allogeneic CAR T therapy, provided the cells are rendered incapable of eliciting graft versus host disease (GvHD). One approach to the production of these cells is gene editing to eliminate expression of the endogenous T cell receptor (TCR). Here we report a streamlined strategy for generating allogeneic CAR T cells by targeting the insertion of a CAR transgene directly into the native TCR locus using an engineered homing endonuclease and an AAV donor template. We demonstrate that anti-CD19 CAR T cells produced in this manner do not express the endogenous TCR, exhibit potent effector functions in vitro, and mediate clearance of CD19+ tumors in an in vivo mouse model.
Collapse
|
244
|
Weinberg MS, Criswell HE, Powell SK, Bhatt AP, McCown TJ. Viral Vector Reprogramming of Adult Resident Striatal Oligodendrocytes into Functional Neurons. Mol Ther 2017; 25:928-934. [PMID: 28202388 DOI: 10.1016/j.ymthe.2017.01.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 01/06/2017] [Accepted: 01/11/2017] [Indexed: 11/18/2022] Open
Abstract
Recent advances suggest that in vivo reprogramming of endogenous cell populations provides a viable alternative for neuron replacement. Astrocytes and oligodendrocyte precursor cells can be induced to transdifferentiate into neurons in the CNS, but, in these instances, reprogramming requires either transgenic mice or retroviral-mediated gene expression. We developed a microRNA (miRNA)-GFP construct that in vitro significantly reduced the expression of polypyrimidine tract-binding protein, and, subsequently, we packaged this construct in a novel oligodendrocyte preferring adeno-associated virus vector. Ten days after rat striatal transduction, the vast majority of the GFP-positive cells were oligodendrocytes, but 6 weeks to 6 months later, the majority of GFP-positive cells exhibited neuronal morphology and co-localized with the neuronal marker NeuN. Patch-clamp studies on striatal slices established that the GFP-positive cells exhibited electrophysiological properties indicative of mature neurons, such as spontaneous action potentials and spontaneous inhibitory postsynaptic currents. Also, 3 months after striatal vector administration, GFP-positive terminals in the ipsilateral globus pallidus or substantia nigra retrogradely transported fluorescent beads back to GFP-positive striatal cell bodies, indicating the presence of functional presynaptic terminals. Thus, this viral vector approach provides a potential means to harness resident oligodendrocytes as an endogenous source for in vivo neuronal replacement.
Collapse
Affiliation(s)
- Marc S Weinberg
- UNC Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hugh E Criswell
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sara K Powell
- UNC Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Aadra P Bhatt
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Thomas J McCown
- UNC Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
245
|
Xiao PJ, Mitchell AM, Huang L, Li C, Samulski RJ. Disruption of Microtubules Post-Virus Entry Enhances Adeno-Associated Virus Vector Transduction. Hum Gene Ther 2016; 27:309-24. [PMID: 26942476 DOI: 10.1089/hum.2016.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Perinuclear retention of viral particles is a poorly understood phenomenon observed during many virus infections. In this study, we investigated whether perinuclear accumulation acts as a barrier to limit recombinant adeno-associated virus (rAAV) transduction. After nocodazole treatment to disrupt microtubules at microtubule-organization center (MT-MTOC) after virus entry, we observed higher rAAV transduction. To elucidate the role of MT-MTOC in rAAV infection and study its underlying mechanisms, we demonstrated that rAAV's perinuclear localization was retained by MT-MTOC with fluorescent analysis, and enhanced rAAV transduction from MT-MTOC disruption was dependent on the rAAV capsid's nuclear import signals. Interestingly, after knocking down RhoA or inhibiting its downstream effectors (ROCK and Actin), MT-MTOC disruption failed to increase rAAV transduction or nuclear entry. These data suggest that enhancement of rAAV transduction is the result of increased trafficking to the nucleus via the RhoA-ROCK-Actin pathway. Ten-fold higher rAAV transduction was also observed by disrupting MT-MTOC in brain, liver, and tumor in vivo. In summary, this study indicates that virus perinuclear accumulation at MT-MTOC is a barrier-limiting parameter for effective rAAV transduction and defines a novel defense mechanism by which host cells restrain viral invasion.
Collapse
Affiliation(s)
- Ping-Jie Xiao
- 1 Gene Therapy Center, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina.,2 Cell and Developmental Biology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina
| | - Angela M Mitchell
- 1 Gene Therapy Center, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina.,3 Department of Microbiology and Immunology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina
| | - Lu Huang
- 4 Department of Statistics, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina
| | - Chengwen Li
- 1 Gene Therapy Center, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina
| | - R Jude Samulski
- 1 Gene Therapy Center, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina.,5 Department of Pharmacology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina
| |
Collapse
|
246
|
Wang L, Luo JY, Li B, Tian XY, Chen LJ, Huang Y, Liu J, Deng D, Lau CW, Wan S, Ai D, Mak KLK, Tong KK, Kwan KM, Wang N, Chiu JJ, Zhu Y, Huang Y. Integrin-YAP/TAZ-JNK cascade mediates atheroprotective effect of unidirectional shear flow. Nature 2016; 540:579-582. [DOI: 10.1038/nature20602] [Citation(s) in RCA: 334] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 11/03/2016] [Indexed: 12/31/2022]
|
247
|
Manzoor F, Johnson MC, Li C, Samulski RJ, Wang B, Tisch R. β-cell-specific IL-35 therapy suppresses ongoing autoimmune diabetes in NOD mice. Eur J Immunol 2016; 47:144-154. [PMID: 27859048 DOI: 10.1002/eji.201646493] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 10/03/2016] [Accepted: 11/03/2016] [Indexed: 12/30/2022]
Abstract
IL-35 is a recently identified cytokine exhibiting potent immunosuppressive properties. The therapeutic potential and effects of IL-35 on pathogenic T effector cells (Teff) and Foxp3+ Treg, however, are ill defined. We tested the capacity of IL-35 to suppress ongoing autoimmunity in NOD mice. For this purpose, an adeno-associated virus vector in which IL-35 transgene expression is selectively targeted to β cells via an insulin promoter (AAV8mIP-IL35) was used. AAV8mIP-IL35 vaccination of NOD mice at a late preclinical stage of type 1 diabetes (T1D) suppressed β-cell autoimmunity and prevented diabetes onset. Numbers of islet-resident conventional CD4+ and CD8+ T cells, and DCs were reduced within 4 weeks of AAV8mIP-IL35 treatment. The diminished islet T-cell pool correlated with suppressed proliferation, and a decreased frequency of IFN-γ-expressing Teff. Ectopic IL-35 also reduced islet Foxp3+ Treg numbers and proliferation, and protection was independent of induction/expansion of adaptive islet immunoregulatory T cells. These findings demonstrate that IL-35-mediated suppression is sufficiently robust to block established β-cell autoimmunity, and support the use of IL-35 to treat T1D and other T-cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Fatima Manzoor
- Department of Microbiology & Immunology, Chapel Hill, NC, USA
| | - Mark C Johnson
- Department of Microbiology & Immunology, Chapel Hill, NC, USA
| | - Chengwen Li
- Gene Therapy Center, Chapel Hill, NC, USA.,Department of Pharmacology, Chapel Hill, NC, USA
| | - R Jude Samulski
- Gene Therapy Center, Chapel Hill, NC, USA.,Department of Pharmacology, Chapel Hill, NC, USA
| | - Bo Wang
- Department of Microbiology & Immunology, Chapel Hill, NC, USA
| | - Roland Tisch
- Department of Microbiology & Immunology, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
248
|
Bernt A, Rangrez AY, Eden M, Jungmann A, Katz S, Rohr C, Müller OJ, Katus HA, Sossalla ST, Williams T, Ritter O, Frank D, Frey N. Sumoylation-independent activation of Calcineurin-NFAT-signaling via SUMO2 mediates cardiomyocyte hypertrophy. Sci Rep 2016; 6:35758. [PMID: 27767176 PMCID: PMC5073337 DOI: 10.1038/srep35758] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 10/03/2016] [Indexed: 12/15/2022] Open
Abstract
The objective of this study was to identify unknown modulators of Calcineurin (Cn)-NFAT signaling. Measurement of NFAT reporter driven luciferase activity was therefore utilized to screen a human cardiac cDNA-library (~107 primary clones) in C2C12 cells through serial dilutions until single clones could be identified. This extensive screening strategy culminated in the identification of SUMO2 as a most efficient Cn-NFAT activator. SUMO2-mediated activation of Cn-NFAT signaling in cardiomyocytes translated into a hypertrophic phenotype. Prohypertrophic effects were also observed in mice expressing SUMO2 in the heart using AAV9 (Adeno-associated virus), complementing the in vitro findings. In addition, increased SUMO2-mediated sumoylation in human cardiomyopathy patients and in mouse models of cardiomyopathy were observed. To decipher the underlying mechanism, we generated a sumoylation-deficient SUMO2 mutant (ΔGG). Surprisingly, ΔGG replicated Cn-NFAT-activation and the prohypertrophic effects of native SUMO2, both in vitro and in vivo, suggesting a sumoylation-independent mechanism. Finally, we discerned a direct interaction between SUMO2 and CnA, which promotes CnA nuclear localization. In conclusion, we identified SUMO2 as a novel activator of Cn-NFAT signaling in cardiomyocytes. In broader terms, these findings reveal an unexpected role for SUMO2 in cardiac hypertrophy and cardiomyopathy, which may open the possibility for therapeutic manipulation of this pathway.
Collapse
Affiliation(s)
- Alexander Bernt
- Dept of Internal Medicine III (Cardiology and Angiology) UKSH, Campus Kiel, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg-Kiel-Lübeck, Kiel, Germany
| | - Ashraf Y Rangrez
- Dept of Internal Medicine III (Cardiology and Angiology) UKSH, Campus Kiel, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg-Kiel-Lübeck, Kiel, Germany
| | - Matthias Eden
- Dept of Internal Medicine III (Cardiology and Angiology) UKSH, Campus Kiel, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg-Kiel-Lübeck, Kiel, Germany
| | - Andreas Jungmann
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg-Kiel-Lübeck, Kiel, Germany.,Dept of Internal Medicine III, University of Heidelberg, Germany
| | - Sylvia Katz
- Dept of Internal Medicine III, University of Heidelberg, Germany
| | - Claudia Rohr
- Dept of Internal Medicine III, University of Heidelberg, Germany
| | - Oliver J Müller
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg-Kiel-Lübeck, Kiel, Germany.,Dept of Internal Medicine III, University of Heidelberg, Germany
| | - Hugo A Katus
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg-Kiel-Lübeck, Kiel, Germany.,Dept of Internal Medicine III, University of Heidelberg, Germany
| | - Samuel T Sossalla
- Dept of Internal Medicine III (Cardiology and Angiology) UKSH, Campus Kiel, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg-Kiel-Lübeck, Kiel, Germany
| | - Tatjana Williams
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg-Kiel-Lübeck, Kiel, Germany.,Dept of Internal Medicine I (Cardiology), University Hospital of Würzburg, Germany
| | - Oliver Ritter
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg-Kiel-Lübeck, Kiel, Germany.,Dept of Internal Medicine I (Cardiology), University Hospital of Würzburg, Germany
| | - Derk Frank
- Dept of Internal Medicine III (Cardiology and Angiology) UKSH, Campus Kiel, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg-Kiel-Lübeck, Kiel, Germany
| | - Norbert Frey
- Dept of Internal Medicine III (Cardiology and Angiology) UKSH, Campus Kiel, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg-Kiel-Lübeck, Kiel, Germany
| |
Collapse
|
249
|
Heparan Sulfate Binding Promotes Accumulation of Intravitreally Delivered Adeno-associated Viral Vectors at the Retina for Enhanced Transduction but Weakly Influences Tropism. J Virol 2016; 90:9878-9888. [PMID: 27558418 DOI: 10.1128/jvi.01568-16] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 08/15/2016] [Indexed: 12/20/2022] Open
Abstract
Many adeno-associated virus (AAV) serotypes efficiently transduce the retina when delivered to the subretinal space but show limited success when delivered to the vitreous due to the inner limiting membrane (ILM). Subretinal delivery of AAV serotype 2 (AAV2) and its heparan sulfate (HS)-binding-deficient capsid led to similar expression, indicating transduction of the outer retina occurred by HS-independent mechanisms. However, intravitreal delivery of HS-ablated recombinant AAV2 (rAAV2) led to a 300-fold decrease in transduction compared to AAV2. Fluorescence in situ hybridization of AAV transgenes was used to identify differences in retinal trafficking and revealed that HS binding was responsible for AAV2 accumulation at the ILM. This mechanism was tested on human ex vivo retinas and showed similar accumulation with HS-binding AAV2 capsid only. To evaluate if HS binding could be applied to other AAV serotypes to enhance their transduction, AAV1 and AAV8 were modified to bind HS with a single-amino-acid mutation and tested in mice. Both HS-binding mutants of AAV1 and AAV8 had higher intravitreal transduction than their non-HS-binding parent capsid due to increased retinal accumulation. To understand the influence that HS binding has on tropism, chimeric AAV2 capsids with dual-glycan usage were tested intravitreally in mice. Compared to HS binding alone, these chimeric capsids displayed enhanced transduction that was correlated with a change in tropism. Taken together, these data indicate that HS binding serves to sequester AAV capsids from the vitreous to the ILM but does not influence retinal tropism. The enhanced retinal transduction of HS-binding capsids provides a rational design strategy for engineering capsids for intravitreal delivery. IMPORTANCE Adeno-associated virus (AAV) has become the vector of choice for viral gene transfer and has shown great promise in clinical trials. The need for development of an easy, less invasive injection route for ocular gene therapy is met by intravitreal delivery, but delivery of AAV by this route results in poor transduction outcomes. The inner limiting membrane (ILM) creates a barrier separating the vitreous and the retina. Binding of AAV to heparan sulfate proteoglycan (HSPG) at the ILM may allow the virus to traverse this barrier for better retinal transduction. We show that HSPG binding is correlated with greater accumulation and penetration of AAV in the retina. We demonstrated that this accumulation is conserved across mouse and human retinas and that the addition of HSPG binding to other AAV capsids can increase the number of vectors accumulating at the ILM without dictating tropism.
Collapse
|
250
|
Chronic Kappa opioid receptor activation modulates NR2B: Implication in treatment resistant depression. Sci Rep 2016; 6:33401. [PMID: 27634008 PMCID: PMC5025743 DOI: 10.1038/srep33401] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/25/2016] [Indexed: 12/15/2022] Open
Abstract
Psychotomimetic and prodepressive effect by kappa opioid receptor (KOR) activation in rodents and human is widely known. Significantly, recent clinical investigations demonstrated the salutary effects of KOR antagonists in patients with treatment resistant depression, indicating essential role of KOR signaling in refractory depression. This study was undertaken to reveal the molecular determinant of KOR mediated depression and antidepressant response of KOR antagonist. We observed that chronic KOR activation by U50488, a selective KOR agonist, significantly increased depression like symptoms (behavioral despair, anhedonia and sociability) in C57BL/6J mice, which were blocked by KOR antagonist norBNI and antidepressant imipramine, but not by fluoxetine or citalopram. Further, chronic KOR activation increased phosphorylation of NR2B subunit of NMDA at tyrosine 1472 (pNR2B NMDA) in the hippocampus, but not in the cortex. Similar to behavioral effects norBNI and imipramine, but not SSRIs, blocked NR2B phosphorylation. Moreover, KOR induced depression like behaviors were reversed by NR2B selective inhibitor Ro 25-6981. Mechanistic studies in primary cultured neurons and brain tissues using genetic and pharmacological approaches revealed that stimulation of KOR modulates several molecular correlates of depression. Thus, these findings elucidate molecular mechanism of KOR signaling in treatment resistant depression like behaviors in mice.
Collapse
|